51
|
Wang Y, Gao J, Zhang L, Yang R, Zhang Y, Shan L, Li X, Ma K. Bioinformatics analysis of lncRNA-related ceRNA networks in the peripheral blood lymphocytes of Kazakh patients with essential hypertension in Xinjiang. Front Cardiovasc Med 2023; 10:1155767. [PMID: 37396592 PMCID: PMC10311024 DOI: 10.3389/fcvm.2023.1155767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/25/2023] [Indexed: 07/04/2023] Open
Abstract
Objective Here, we aimed to investigate long non-coding RNA (lncRNA) expression characteristics in the peripheral blood lymphocytes of Xinjiang Kazakh people with essential hypertension and the underlying regulatory mechanisms of competing endogenous RNAs (ceRNA). Methods From April 2016 to May 2019, six Kazakh patients with essential hypertension and six Kazakh healthy participants were randomly selected from the inpatient and outpatient cardiology departments of the First Affiliated Hospital of Shihezi University Medical College, Xinjiang. After detecting the expression levels of lncRNA and mRNA in the peripheral blood lymphocytes using gene chip technology, their levels in the hypertensive group were compared with those in the control group. Six differentially expressed lncRNAs were randomly selected for real-time PCR to verify the accuracy and reliability of the gene chip results. GO functional clustering and KEGG pathway analyses were performed for differentially expressed genes. The ceRNA regulatory network of lncRNA-miRNA-mRNA was constructed, followed by visualization of the results. The expressions of miR-139-5p and DCBLD2 after PVT1 overexpression in 293T cells were detected by qRT-PCR and Western blot. Results In the test group, 396 and 511 differentially expressed lncRNAs and mRNAs, respectively, were screened out. The trend of real-time PCR results was consistent with that of the microarray results. The differentially expressed mRNAs were found to be primarily involved in the adhesion spot, leukocyte migration via endothelial cells, gap junction, actin cytoskeleton regulation, and extracellular matrix-receptor interaction signaling pathways. By constructing the ceRNA regulatory network, we found that lncRNA PVT1-miR-139-5p-DCBLD2 has a potential ceRNA regulatory mechanism involved in the development of essential hypertension in Xinjiang Kazakh people. In 293T cells, lncRNA PVT1 overexpression inhibited miR-139-5p and DCBLD2 levels. Conclusions Our findings indicate that differentially expressed lncRNAs may be involved in the development of essential hypertension. lncRNA PVT1-miR-139-5p-DCBLD2 was indicated to comprise a potential ceRNA regulatory mechanism involved in the development of essential hypertension in the Xinjiang Kazakh population. Thus, it may act as a novel screening marker or therapeutic target for essential hypertension in this population.
Collapse
Affiliation(s)
- Yan Wang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China
| | - Jie Gao
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi, China
| | - Liang Zhang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi, China
| | - Rui Yang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi, China
| | - Yingying Zhang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi, China
| | - Liya Shan
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi, China
| | - Xinzhi Li
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China
- Department of Pathophysiology, Shihezi University School of Medicine, Shihezi, China
| | - Ketao Ma
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi, China
| |
Collapse
|
52
|
Bernabei I, Hansen U, Ehirchiou D, Brinckmann J, Chobaz V, Busso N, Nasi S. CD11b Deficiency Favors Cartilage Calcification via Increased Matrix Vesicles, Apoptosis, and Lysyl Oxidase Activity. Int J Mol Sci 2023; 24:ijms24119776. [PMID: 37298730 DOI: 10.3390/ijms24119776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/31/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Pathological cartilage calcification is a hallmark feature of osteoarthritis, a common degenerative joint disease, characterized by cartilage damage, progressively causing pain and loss of movement. The integrin subunit CD11b was shown to play a protective role against cartilage calcification in a mouse model of surgery-induced OA. Here, we investigated the possible mechanism by which CD11b deficiency could favor cartilage calcification by using naïve mice. First, we found by transmission electron microscopy (TEM) that CD11b KO cartilage from young mice presented early calcification spots compared with WT. CD11b KO cartilage from old mice showed progression of calcification areas. Mechanistically, we found more calcification-competent matrix vesicles and more apoptosis in both cartilage and chondrocytes isolated from CD11b-deficient mice. Additionally, the extracellular matrix from cartilage lacking the integrin was dysregulated with increased collagen fibrils with smaller diameters. Moreover, we revealed by TEM that CD11b KO cartilage had increased expression of lysyl oxidase (LOX), the enzyme that catalyzes matrix crosslinks. We confirmed this in murine primary CD11b KO chondrocytes, where Lox gene expression and crosslinking activity were increased. Overall, our results suggest that CD11b integrin regulates cartilage calcification through reduced MV release, apoptosis, LOX activity, and matrix crosslinking. As such, CD11b activation might be a key pathway for maintaining cartilage integrity.
Collapse
Affiliation(s)
- Ilaria Bernabei
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Uwe Hansen
- Institute for Musculoskeletal Medicine, University Hospital of Münster, 48149 Münster, Germany
| | - Driss Ehirchiou
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Jürgen Brinckmann
- Department of Dermatology, University of Lübeck, 23562 Lübeck, Germany
| | - Veronique Chobaz
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Nathalie Busso
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Sonia Nasi
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital, 1011 Lausanne, Switzerland
| |
Collapse
|
53
|
Sanati M, Afshari AR, Aminyavari S, Kesharwani P, Jamialahmadi T, Sahebkar A. RGD-engineered nanoparticles as an innovative drug delivery system in cancer therapy. J Drug Deliv Sci Technol 2023; 84:104562. [DOI: 10.1016/j.jddst.2023.104562] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
|
54
|
Liu J, Lu F, Ithychanda SS, Apostol M, Das M, Deshpande G, Plow EF, Qin J. A mechanism of platelet integrin αIIbβ3 outside-in signaling through a novel integrin αIIb subunit-filamin-actin linkage. Blood 2023; 141:2629-2641. [PMID: 36867840 PMCID: PMC10356577 DOI: 10.1182/blood.2022018333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 02/24/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023] Open
Abstract
The communication of talin-activated integrin αIIbβ3 with the cytoskeleton (integrin outside-in signaling) is essential for platelet aggregation, wound healing, and hemostasis. Filamin, a large actin crosslinker and integrin binding partner critical for cell spreading and migration, is implicated as a key regulator of integrin outside-in signaling. However, the current dogma is that filamin, which stabilizes inactive αIIbβ3, is displaced from αIIbβ3 by talin to promote the integrin activation (inside-out signaling), and how filamin further functions remains unresolved. Here, we show that while associating with the inactive αIIbβ3, filamin also associates with the talin-bound active αIIbβ3 to mediate platelet spreading. Fluorescence resonance energy transfer-based analysis reveals that while associating with both αIIb and β3 cytoplasmic tails (CTs) to maintain the inactive αIIbβ3, filamin is spatiotemporally rearranged to associate with αIIb CT alone on activated αIIbβ3. Consistently, confocal cell imaging indicates that integrin α CT-linked filamin gradually delocalizes from the β CT-linked focal adhesion marker-vinculin likely because of the separation of integrin α/β CTs occurring during integrin activation. High-resolution crystal and nuclear magnetic resonance structure determinations unravel that the activated integrin αIIb CT binds to filamin via a striking α-helix→β-strand transition with a strengthened affinity that is dependent on the integrin-activating membrane environment containing enriched phosphatidylinositol 4,5-bisphosphate. These data suggest a novel integrin αIIb CT-filamin-actin linkage that promotes integrin outside-in signaling. Consistently, disruption of such linkage impairs the activation state of αIIbβ3, phosphorylation of focal adhesion kinase/proto-oncogene tyrosine kinase Src, and cell migration. Together, our findings advance the fundamental understanding of integrin outside-in signaling with broad implications in blood physiology and pathology.
Collapse
Affiliation(s)
- Jianmin Liu
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Fan Lu
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH
| | - Sujay Subbayya Ithychanda
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Marcin Apostol
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Mitali Das
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Gauravi Deshpande
- Imaging Core, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Edward F. Plow
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Jun Qin
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
55
|
Tang C, Liu M, Zhou Z, Li H, Yang C, Yang L, Xiang J. Treadmill Exercise Alleviates Cognition Disorder by Activating the FNDC5: Dual Role of Integrin αV/β5 in Parkinson's Disease. Int J Mol Sci 2023; 24:ijms24097830. [PMID: 37175535 PMCID: PMC10178565 DOI: 10.3390/ijms24097830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/13/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
Parkinson's disease with cognitive impairment (PD-CI) results in several clinical outcomes for which specific treatment is lacking. Although the pathogenesis of PD-CI has not yet been fully elucidated, it is related to neuronal plasticity decline in the hippocampus region. The dopaminergic projections from the substantia nigra to the hippocampus are critical in regulating hippocampal plasticity. Recently, aerobic exercise has been recognized as an effective therapeutic strategy for enhancing plasticity through the secretion of various muscle factors. The exact role of FNDC5-an upregulated, newly identified myokine produced after exercise-in mediating hippocampal plasticity and regional dopaminergic projections in PD-CI remains unclear. In this study, the effect of treadmill exercise on hippocampal synaptic plasticity was evaluated in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced chronic PD models. The results showed that treadmill exercise substantially alleviated the motor dysfunction, cognition disorder, and dopaminergic neuron degeneration induced by MPTP. Here, we discovered that the quadriceps, serum, and brain FNDC5 levels were lower in PD mice and that intervention with treadmill exercise restored FNDC5 levels. Moreover, treadmill exercise enhanced the synaptic plasticity of hippocampal pyramidal neurons via increased dopamine levels and BDNF in the PD mice. The direct protective effect of FNDC5 is achieved by promoting the secretion of BDNF in the hippocampal neurons via binding the integrin αVβ5 receptor, thereby improving synaptic plasticity. Regarding the indirect protection effect, FNDC5 promotes the dopaminergic connection from the substantia nigra to the hippocampus by mediating the interaction between the integrin αVβ5 of the hippocampal neurons and the CD90 molecules on the membrane of dopaminergic terminals. Our findings demonstrated that treadmill exercise could effectively alleviate cognitive disorders via the activation of the FNDC5-BDNF pathway and enhance the dopaminergic synaptic connection from SNpc to the hippocampus in the MPTP-induced chronic PD model.
Collapse
Affiliation(s)
- Chuanxi Tang
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology, Xuzhou Medical University, Xuzhou 221004, China
| | - Mengting Liu
- Department of Rehabilitation, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002, China
- The College of Medical Technology, Xuzhou Medical University, Xuzhou 221004, China
| | - Zihang Zhou
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology, Xuzhou Medical University, Xuzhou 221004, China
| | - Hao Li
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology, Xuzhou Medical University, Xuzhou 221004, China
| | - Chenglin Yang
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology, Xuzhou Medical University, Xuzhou 221004, China
| | - Li Yang
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology, Xuzhou Medical University, Xuzhou 221004, China
| | - Jie Xiang
- Department of Rehabilitation, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002, China
| |
Collapse
|
56
|
Jeruzalska E, Mazur AJ. The Role of non-muscle actin paralogs in cell cycle progression and proliferation. Eur J Cell Biol 2023; 102:151315. [PMID: 37099935 DOI: 10.1016/j.ejcb.2023.151315] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 04/14/2023] [Accepted: 04/17/2023] [Indexed: 04/28/2023] Open
Abstract
Uncontrolled cell proliferation leads to several pathologies, including cancer. Thus, this process must be tightly regulated. The cell cycle accounts for cell proliferation, and its progression is coordinated with changes in cell shape, for which cytoskeleton reorganization is responsible. Rearrangement of the cytoskeleton allows for its participation in the precise division of genetic material and cytokinesis. One of the main cytoskeletal components is filamentous actin-based structures. Mammalian cells have at least six actin paralogs, four of which are muscle-specific, while two, named β- and γ-actin, are abundantly present in all types of cells. This review summarizes the findings that establish the role of non-muscle actin paralogs in regulating cell cycle progression and proliferation. We discuss studies showing that the level of a given non-muscle actin paralog in a cell influences the cell's ability to progress through the cell cycle and, thus, proliferation. Moreover, we elaborate on the non-muscle actins' role in regulating gene transcription, interactions of actin paralogs with proteins involved in controlling cell proliferation, and the contribution of non-muscle actins to different structures in a dividing cell. The data cited in this review show that non-muscle actins regulate the cell cycle and proliferation through varying mechanisms. We point to the need for further studies addressing these mechanisms.
Collapse
Affiliation(s)
- Estera Jeruzalska
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Poland
| | - Antonina J Mazur
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Poland.
| |
Collapse
|
57
|
Abboud Asleh M, Zaher M, Asleh J, Jadon J, Shaulov L, Yelin R, Schultheiss TM. A morphogenetic wave in the chick embryo lateral mesoderm generates mesenchymal-epithelial transition through a 3D-rosette intermediate. Dev Cell 2023:S1534-5807(23)00133-8. [PMID: 37080204 DOI: 10.1016/j.devcel.2023.03.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 02/24/2023] [Accepted: 03/24/2023] [Indexed: 04/22/2023]
Abstract
Formation of epithelia through mesenchymal-epithelial transition (MET) is essential for embryonic development and for many physiological and pathological processes. This study investigates MET in vivo in the chick embryo lateral mesoderm, where a multilayered mesenchyme transforms into two parallel epithelial sheets that constitute the coelomic lining of the embryonic body cavity. Prior to MET initiation, mesenchymal cells exhibit non-polarized distribution of multiple polarity markers, albeit not aPKC. We identified an epithelializing wave that sweeps across the lateral mesoderm, the wavefront of which is characterized by the accumulation of basal fibronectin and a network of 3D rosettes composed of polarized, wedge-shaped cells surrounding a central focus of apical markers, now including aPKC. Initiation of the MET process is dependent on extracellular matrix-integrin signaling acting through focal adhesion kinase and talin, whereas progression through the rosette phase requires aPKC function. We present a stepwise model for MET, comprising polarization, 3D-rosette, and epithelialization stages.
Collapse
Affiliation(s)
- Manar Abboud Asleh
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Mira Zaher
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Jad Asleh
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Julian Jadon
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Lihi Shaulov
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Ronit Yelin
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Thomas M Schultheiss
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel.
| |
Collapse
|
58
|
Dey H, Vasudevan K, Doss C. GP, Kumar SU, El Allali A, Alsamman AM, Zayed H. Integrated gene network analysis sheds light on understanding the progression of Osteosarcoma. Front Med (Lausanne) 2023; 10:1154417. [PMID: 37081847 PMCID: PMC10110863 DOI: 10.3389/fmed.2023.1154417] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/20/2023] [Indexed: 04/07/2023] Open
Abstract
Introduction Osteosarcoma is a rare disorder among cancer, but the most frequently occurring among sarcomas in children and adolescents. It has been reported to possess the relapsing capability as well as accompanying collateral adverse effects which hinder the development process of an effective treatment plan. Using networks of omics data to identify cancer biomarkers could revolutionize the field in understanding the cancer. Cancer biomarkers and the molecular mechanisms behind it can both be understood by studying the biological networks underpinning the etiology of the disease. Methods In our study, we aimed to highlight the hub genes involved in gene-gene interaction network to understand their interaction and how they affect the various biological processes and signaling pathways involved in Osteosarcoma. Gene interaction network provides a comprehensive overview of functional gene analysis by providing insight into how genes cooperatively interact to elicit a response. Because gene interaction networks serve as a nexus to many biological problems, their employment of it to identify the hub genes that can serve as potential biomarkers remain widely unexplored. A dynamic framework provides a clear understanding of biological complexity and a pathway from the gene level to interaction networks. Results Our study revealed various hub genes viz. TP53, CCND1, CDK4, STAT3, and VEGFA by analyzing various topological parameters of the network, such as highest number of interactions, average shortest path length, high cluster density, etc. Their involvement in key signaling pathways, such as the FOXM1 transcription factor network, FAK-mediated signaling events, and the ATM pathway, makes them significant candidates for studying the disease. The study also highlighted significant enrichment in GO terms (Biological Processes, Molecular Function, and Cellular Processes), such as cell cycle signal transduction, cell communication, kinase binding, transcription factor activity, nucleoplasm, PML body, nuclear body, etc. Conclusion To develop better therapeutics, a specific approach toward the disease targeting the hub genes involved in various signaling pathways must have opted to unravel the complexity of the disease. Our study has highlighted the candidate hub genes viz. TP53, CCND1 CDK4, STAT3, VEGFA. Their involvement in the major signaling pathways of Osteosarcoma makes them potential candidates to be targeted for drug development. The highly enriched signaling pathways include FOXM1 transcription pathway, ATM signal-ling pathway, FAK mediated signaling events, Arf6 signaling events, mTOR signaling pathway, and Integrin family cell surface interactions. Targeting the hub genes and their associated functional partners which we have reported in our studies may be efficacious in developing novel therapeutic targets.
Collapse
Affiliation(s)
- Hrituraj Dey
- Department of Biotechnology, School of Applied Sciences, REVA University, Bangalore, India
| | - Karthick Vasudevan
- Department of Biotechnology, School of Applied Sciences, REVA University, Bangalore, India
| | - George Priya Doss C.
- Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore, India
| | - S. Udhaya Kumar
- Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore, India
| | - Achraf El Allali
- African Genome Center, Mohammed VI Polytechnic University, Ben Guerir, Morocco
| | - Alsamman M. Alsamman
- Agriculture Genetic Engineering Research Institute (AGERI), Agriculture Research Center (ARC), Giza, Egypt
- International Center for Agricultural Research in the Dry Areas (ICARDA), Giza, Egypt
| | - Hatem Zayed
- Department of Biomedical Sciences College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
59
|
Chu PH, Chen SC, Chen HY, Wu CB, Huang WT, Chiang HY. Astrocyte-associated fibronectin promotes the proinflammatory phenotype of astrocytes through β1 integrin activation. Mol Cell Neurosci 2023; 125:103848. [PMID: 36948232 DOI: 10.1016/j.mcn.2023.103848] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/24/2023] Open
Abstract
Astrocytes are key players in neuroinflammation. In response to central nervous system (CNS) injury or disease, astrocytes undergo reactive astrogliosis, which is characterized by increased proliferation, migration, and glial fibrillary acidic protein (GFAP) expression. Activation of the transcription factor nuclear factor-κB (NF-κB) and upregulation of downstream proinflammatory mediators in reactive astrocytes induce a proinflammatory phenotype in astrocytes, thereby exacerbating neuroinflammation by establishing an inflammatory loop. In this study, we hypothesized that excessive fibronectin (FN) derived from reactive astrocytes would induce this proinflammatory phenotype in astrocytes in an autocrine manner. We exogenously treated astrocytes with monomer FN, which can be incorporated into the extracellular matrix (ECM), to mimic plasma FN extravasated through a compromised blood-brain barrier in neuroinflammation. We also induced de novo synthesis and accumulation of astrocyte-derived FN through tumor necrosis factor-α (TNF-α) stimulation. The excessive FN deposition resulting from both treatments initiated reactive astrogliosis and triggered NF-κB signaling in the cultured astrocytes. In addition, inhibition of FN accumulation in the ECM by the FN inhibitor pUR4 strongly attenuated the FN- and TNF-α-induced GFAP expression, NF-κB activation, and proinflammatory mediator production of astrocytes by interrupting FN-β1 integrin coupling and thus the inflammatory loop. In an in vivo experiment, intrathecal injection of pUR4 considerably ameliorated FN deposition, GFAP expression, and NF-κB activation in inflamed spinal cord, suggesting the therapeutic potential of pUR4 for attenuating neuroinflammation and promoting neuronal function restoration.
Collapse
Affiliation(s)
- Pao-Hsien Chu
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan; Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Shao-Chi Chen
- Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsin-Yung Chen
- Department of Occupational Therapy, Graduate Institute of Behavioral Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Cheng-Bei Wu
- Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wei-Ting Huang
- Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hou-Yu Chiang
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan; Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
60
|
Genduso S, Freytag V, Schetler D, Kirchner L, Schiecke A, Maar H, Wicklein D, Gebauer F, Bröker K, Stürken C, Milde-Langosch K, Oliveira-Ferrer L, Ricklefs FL, Ewald F, Wolters-Eisfeld G, Riecken K, Unrau L, Krause L, Bohnenberger H, Offermann A, Perner S, Sebens S, Lamszus K, Diehl L, Linder S, Jücker M, Schumacher U, Lange T. Tumor cell integrin β4 and tumor stroma E-/P-selectin cooperatively regulate tumor growth in vivo. J Hematol Oncol 2023; 16:23. [PMID: 36932441 PMCID: PMC10022201 DOI: 10.1186/s13045-023-01413-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 02/13/2023] [Indexed: 03/19/2023] Open
Abstract
BACKGROUND The immunological composition of the tumor microenvironment has a decisive influence on the biological course of cancer and is therefore of profound clinical relevance. In this study, we analyzed the cooperative effects of integrin β4 (ITGB4) on tumor cells and E-/P-selectin on endothelial cells within the tumor stroma for regulating tumor growth by shaping the local and systemic immune environment. METHODS We used several preclinical mouse models for different solid human cancer types (xenograft and syngeneic) to explore the role of ITGB4 (shRNA-mediated knockdown in tumor cells) and E-/P-selectins (knockout in mice) for tumor growth; effects on apoptosis, proliferation and intratumoral signaling pathways were determined by histological and biochemical methods and 3D in vitro experiments; changes in the intratumoral and systemic immune cell composition were determined by flow cytometry and immunohistochemistry; chemokine levels and their attracting potential were measured by ELISA and 3D invasion assays. RESULTS We observed a very robust synergism between ITGB4 and E-/P-selectin for the regulation of tumor growth, accompanied by an increased recruitment of CD11b+ Gr-1Hi cells with low granularity (i.e., myeloid-derived suppressor cells, MDSCs) specifically into ITGB4-depleted tumors. ITGB4-depleted tumors undergo apoptosis and actively attract MDSCs, well-known to promote tumor growth in several cancers, via increased secretion of different chemokines. MDSC trafficking into tumors crucially depends on E-/P-selectin expression. Analyses of clinical samples confirmed an inverse relationship between ITGB4 expression in tumors and number of tumor-infiltrating leukocytes. CONCLUSIONS These findings suggest a distinct vulnerability of ITGB4Lo tumors for MDSC-directed immunotherapies.
Collapse
Affiliation(s)
- Sandra Genduso
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Vera Freytag
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Daniela Schetler
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Lennart Kirchner
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Alina Schiecke
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Hanna Maar
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
- Institute of Anatomy I, Cancer Center Central Germany, Jena University Hospital, Teichgraben 7, 07743, Jena, Germany
| | - Daniel Wicklein
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
- Department of Anatomy and Cell Biology, University of Marburg, Robert-Koch-Strasse 8, 35037, Marburg, Germany
| | - Florian Gebauer
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Hospital Cologne, Kerpener Strasse 62, 50937, Cologne, Germany
| | - Katharina Bröker
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Christine Stürken
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
- Faculty of Medicine, MSH Medical School Hamburg, Medical University, 20251, Hamburg, Germany
| | - Karin Milde-Langosch
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Franz L Ricklefs
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Florian Ewald
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gerrit Wolters-Eisfeld
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Pediatric Hematology and Oncology, Research Institute Childrens' Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kristoffer Riecken
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ludmilla Unrau
- Institue of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Linda Krause
- Institute of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hanibal Bohnenberger
- Institute of Pathology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075, Göttingen, Germany
| | - Anne Offermann
- Institute of Pathology, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Sven Perner
- Institute of Pathology, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Susanne Sebens
- Institute for Experimental Cancer Research, Kiel University (CAU) and University Medical Center Schleswig-Holstein, Campus Kiel, Arnold-Heller-Strasse 3, 24105, Kiel, Germany
| | - Katrin Lamszus
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Linda Diehl
- Institue of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Linder
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
- Medical School Berlin, Leipziger Platz 10, 10117, Berlin, Germany
| | - Tobias Lange
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
- Institute of Anatomy I, Cancer Center Central Germany, Jena University Hospital, Teichgraben 7, 07743, Jena, Germany.
| |
Collapse
|
61
|
Amiri B, Heyn JCJ, Schreiber C, Rädler JO, Falcke M. On multistability and constitutive relations of cell motion on fibronectin lanes. Biophys J 2023; 122:753-766. [PMID: 36739476 PMCID: PMC10027452 DOI: 10.1016/j.bpj.2023.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/12/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Cell motility on flat substrates exhibits coexisting steady and oscillatory morphodynamics, the biphasic adhesion-velocity relation, and the universal correlation between speed and persistence (UCSP) as simultaneous observations common to many cell types. Their universality and concurrency suggest a unifying mechanism causing all three of them. Stick-slip models for cells on one-dimensional lanes suggest multistability to arise from the nonlinear friction of retrograde flow. This study suggests a mechanical mechanism controlled by integrin signaling on the basis of a biophysical model and analysis of trajectories of MDA-MB-231 cells on fibronectin lanes, which additionally explains the constitutive relations. The experiments exhibit cells with steady or oscillatory morphodynamics and either spread or moving with spontaneous transitions between the dynamic regimes, spread and moving, and spontaneous direction reversals. Our biophysical model is based on the force balance at the protrusion edge, the noisy clutch of retrograde flow, and a response function of friction and membrane drag to integrin signaling. The theory reproduces the experimentally observed cell states, characteristics of oscillations, and state probabilities. Analysis of experiments with the biophysical model establishes a stick-slip oscillation mechanism, and explains multistability of cell states and the statistics of state transitions. It suggests protrusion competition to cause direction reversal events, the statistics of which explain the UCSP. The effect of integrin signaling on drag and friction explains the adhesion-velocity relation and cell behavior at fibronectin density steps. The dynamics of our mechanism are nonlinear flow mechanics driven by F-actin polymerization and shaped by the noisy clutch of retrograde flow friction, protrusion competition via membrane tension, and drag forces. Integrin signaling controls the parameters of the mechanical system.
Collapse
Affiliation(s)
- Behnam Amiri
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Johannes C J Heyn
- Fakultät für Physik, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Christoph Schreiber
- Fakultät für Physik, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Joachim O Rädler
- Fakultät für Physik, Ludwig-Maximilians-Universität München (LMU), Munich, Germany.
| | - Martin Falcke
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; Department of Physics, Humboldt University, Berlin, Germany.
| |
Collapse
|
62
|
Abstract
Non-muscle myosin 2 (NM2) motors are the major contractile machines in most cell types. Unsurprisingly, these ubiquitously expressed actin-based motors power a plethora of subcellular, cellular and multicellular processes. In this Cell Science at a Glance article and the accompanying poster, we review the biochemical properties and mechanisms of regulation of this myosin. We highlight the central role of NM2 in multiple fundamental cellular processes, which include cell migration, cytokinesis, epithelial barrier function and tissue morphogenesis. In addition, we highlight recent studies using advanced imaging technologies that have revealed aspects of NM2 assembly hitherto inaccessible. This article will hopefully appeal to both cytoskeletal enthusiasts and investigators from outside the cytoskeleton field who have interests in one of the many basic cellular processes requiring actomyosin force production.
Collapse
Affiliation(s)
- Melissa A. Quintanilla
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60525, USA
| | - John A. Hammer
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jordan R. Beach
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60525, USA
| |
Collapse
|
63
|
Norris EG, Pan XS, Hocking DC. Receptor-binding domain of SARS-CoV-2 is a functional αv-integrin agonist. J Biol Chem 2023; 299:102922. [PMID: 36669646 PMCID: PMC9846890 DOI: 10.1016/j.jbc.2023.102922] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
Among the novel mutations distinguishing SARS-CoV-2 from similar coronaviruses is a K403R substitution in the receptor-binding domain (RBD) of the viral spike (S) protein within its S1 region. This amino acid substitution occurs near the angiotensin-converting enzyme 2-binding interface and gives rise to a canonical RGD adhesion motif that is often found in native extracellular matrix proteins, including fibronectin. Here, the ability of recombinant S1-RBD to bind to cell surface integrins and trigger downstream signaling pathways was assessed and compared with RGD-containing, integrin-binding fragments of fibronectin. We determined that S1-RBD supported adhesion of fibronectin-null mouse embryonic fibroblasts as well as primary human small airway epithelial cells, while RBD-coated microparticles attached to epithelial monolayers in a cation-dependent manner. Cell adhesion to S1-RBD was RGD dependent and inhibited by blocking antibodies against αv and β3 but not α5 or β1 integrins. Similarly, we observed direct binding of S1-RBD to recombinant human αvβ3 and αvβ6 integrins, but not α5β1 integrins, using surface plasmon resonance. S1-RBD adhesion initiated cell spreading, focal adhesion formation, and actin stress fiber organization to a similar extent as fibronectin. Moreover, S1-RBD stimulated tyrosine phosphorylation of the adhesion mediators FAK, Src, and paxillin; triggered Akt activation; and supported cell proliferation. Thus, the RGD sequence of S1-RBD can function as an αv-selective integrin agonist. This study provides evidence that cell surface αv-containing integrins can respond functionally to spike protein and raises the possibility that S1-mediated dysregulation of extracellular matrix dynamics may contribute to the pathogenesis and/or post-acute sequelae of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Emma G Norris
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Xuan Sabrina Pan
- Department of Biomedical Engineering, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Denise C Hocking
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA; Department of Biomedical Engineering, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA.
| |
Collapse
|
64
|
Paulus J, Nachtigall B, Meyer P, Sewald N. RGD Peptidomimetic MMAE-Conjugate Addressing Integrin αVβ3-Expressing Cells with High Targeting Index. Chemistry 2023; 29:e202203476. [PMID: 36454662 DOI: 10.1002/chem.202203476] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/01/2022] [Accepted: 12/01/2022] [Indexed: 12/03/2022]
Abstract
Small molecule-drug conjugates (SMDCs) mimicking the RGD sequence (-Arg-Gly-Asp-) with a non-peptide moiety require a pharmacophore-independent attachment site. A library of 36 sulfonamide-modified RGD mimetics with nM to pM affinity for integrin αV β3 was synthesized and analysed via DAD mapping. The best structure of the conjugable RGD mimetic was used and a linker was attached to an aromatic ring by Negishi cross-coupling. The product retained high affinity and selectivity for integrin αV β3 . The conjugable RGD mimetic was then attached to an enzymatically cleavable GKGEVA linker equipped with a self-immolative PABC and the antimitotic drug monomethyl auristatin E (MMAE). The resulting SMDC preferred binding to integrin αV β3 over α5 β1 in a ratio of 1 : 4519 (ELISA) and showed selectivity for αV β3 -positive WM115 cells over αV β3 -negative M21-L cells in the in vitro cell adhesion assay as well as in cell viability assays with a targeting index of 134 (M21-L/WM115).
Collapse
Affiliation(s)
- Jannik Paulus
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Beate Nachtigall
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Peter Meyer
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Norbert Sewald
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| |
Collapse
|
65
|
Biose IJ, Ismael S, Ouvrier B, White AL, Bix GJ. The Potential Role of Integrin Signaling in Memory and Cognitive Impairment. Biomolecules 2023; 13:biom13010108. [PMID: 36671492 PMCID: PMC9855855 DOI: 10.3390/biom13010108] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/29/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023] Open
Abstract
Dementia currently has no cure and, due to the increased prevalence and associated economic and personal burden of this condition, current research efforts for the development of potential therapies have intensified. Recently, targeting integrins as a strategy to ameliorate dementia and other forms of cognitive impairment has begun to gain traction. Integrins are major bidirectional signaling receptors in mammalian cells, mediating various physiological processes such as cell-cell interaction and cell adhesion, and are also known to bind to the extracellular matrix. In particular, integrins play a critical role in the synaptic transmission of signals, hence their potential contribution to memory formation and significance in cognitive impairment. In this review, we describe the physiological roles that integrins play in the blood-brain barrier (BBB) and in the formation of memories. We also provide a clear overview of how integrins are implicated in BBB disruption following cerebral pathology. Given that vascular contributions to cognitive impairment and dementia and Alzheimer's' disease are prominent forms of dementia that involve BBB disruption, as well as chronic inflammation, we present current approaches shown to improve dementia-like conditions with integrins as a central focus. We conclude that integrins are vital in memory formation and that their disruption could lead to various forms of cognitive impairment. While further research to understand the relationships between integrins and memory is needed, we propose that the translational relevance of research efforts in this area could be improved through the use of appropriately aged, comorbid, male and female animals.
Collapse
Affiliation(s)
- Ifechukwude Joachim Biose
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Saifudeen Ismael
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Blake Ouvrier
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA 70112, USA
| | - Amanda Louise White
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA 70112, USA
| | - Gregory Jaye Bix
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
- School of Medicine, Tulane University, New Orleans, LA 70112, USA
- Department of Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70122, USA
- Correspondence: ; Tel.: +1-504-988-3564
| |
Collapse
|
66
|
Pang X, He X, Qiu Z, Zhang H, Xie R, Liu Z, Gu Y, Zhao N, Xiang Q, Cui Y. Targeting integrin pathways: mechanisms and advances in therapy. Signal Transduct Target Ther 2023; 8:1. [PMID: 36588107 PMCID: PMC9805914 DOI: 10.1038/s41392-022-01259-6] [Citation(s) in RCA: 427] [Impact Index Per Article: 213.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/14/2022] [Accepted: 11/21/2022] [Indexed: 01/03/2023] Open
Abstract
Integrins are considered the main cell-adhesion transmembrane receptors that play multifaceted roles as extracellular matrix (ECM)-cytoskeletal linkers and transducers in biochemical and mechanical signals between cells and their environment in a wide range of states in health and diseases. Integrin functions are dependable on a delicate balance between active and inactive status via multiple mechanisms, including protein-protein interactions, conformational changes, and trafficking. Due to their exposure on the cell surface and sensitivity to the molecular blockade, integrins have been investigated as pharmacological targets for nearly 40 years, but given the complexity of integrins and sometimes opposite characteristics, targeting integrin therapeutics has been a challenge. To date, only seven drugs targeting integrins have been successfully marketed, including abciximab, eptifibatide, tirofiban, natalizumab, vedolizumab, lifitegrast, and carotegrast. Currently, there are approximately 90 kinds of integrin-based therapeutic drugs or imaging agents in clinical studies, including small molecules, antibodies, synthetic mimic peptides, antibody-drug conjugates (ADCs), chimeric antigen receptor (CAR) T-cell therapy, imaging agents, etc. A serious lesson from past integrin drug discovery and research efforts is that successes rely on both a deep understanding of integrin-regulatory mechanisms and unmet clinical needs. Herein, we provide a systematic and complete review of all integrin family members and integrin-mediated downstream signal transduction to highlight ongoing efforts to develop new therapies/diagnoses from bench to clinic. In addition, we further discuss the trend of drug development, how to improve the success rate of clinical trials targeting integrin therapies, and the key points for clinical research, basic research, and translational research.
Collapse
Affiliation(s)
- Xiaocong Pang
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Xu He
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Zhiwei Qiu
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Hanxu Zhang
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Ran Xie
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Zhiyan Liu
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Yanlun Gu
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Nan Zhao
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Qian Xiang
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034, Beijing, China. .,Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191, Beijing, China.
| | - Yimin Cui
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034, Beijing, China. .,Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191, Beijing, China.
| |
Collapse
|
67
|
Wang K, Wen D, Xu X, Zhao R, Jiang F, Yuan S, Zhang Y, Gao Y, Li Q. Extracellular matrix stiffness-The central cue for skin fibrosis. Front Mol Biosci 2023; 10:1132353. [PMID: 36968277 PMCID: PMC10031116 DOI: 10.3389/fmolb.2023.1132353] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 02/20/2023] [Indexed: 03/29/2023] Open
Abstract
Skin fibrosis is a physiopathological process featuring the excessive deposition of extracellular matrix (ECM), which is the main architecture that provides structural support and constitutes the microenvironment for various cellular behaviors. Recently, increasing interest has been drawn to the relationship between the mechanical properties of the ECM and the initiation and modulation of skin fibrosis, with the engagement of a complex network of signaling pathways, the activation of mechanosensitive proteins, and changes in immunoregulation and metabolism. Simultaneous with the progression of skin fibrosis, the stiffness of ECM increases, which in turn perturbs mechanical and humoral homeostasis to drive cell fate toward an outcome that maintains and enhances the fibrosis process, thus forming a pro-fibrotic "positive feedback loop". In this review, we highlighted the central role of the ECM and its dynamic changes at both the molecular and cellular levels in skin fibrosis. We paid special attention to signaling pathways regulated by mechanical cues in ECM remodeling. We also systematically summarized antifibrotic interventions targeting the ECM, hopefully enlightening new strategies for fibrotic diseases.
Collapse
Affiliation(s)
- Kang Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dongsheng Wen
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuewen Xu
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rui Zhao
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Feipeng Jiang
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Shengqin Yuan
- School of Public Administration, Sichuan University, Chengdu, Sichuan, China
| | - Yifan Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yifan Zhang, ; Ya Gao, ; Qingfeng Li,
| | - Ya Gao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yifan Zhang, ; Ya Gao, ; Qingfeng Li,
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yifan Zhang, ; Ya Gao, ; Qingfeng Li,
| |
Collapse
|
68
|
Takeshita N, Takano-Yamamoto T. Analysis of Chemotactic Property of CCN2/CTGF in Intramembranous Osteogenesis. Methods Mol Biol 2023; 2582:237-253. [PMID: 36370354 DOI: 10.1007/978-1-0716-2744-0_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Chemotaxis is a directed migration of cells in response to a gradient of extracellular molecules called chemoattractants. Development, growth, remodeling, and fracture healing of bones are advanced through intramembranous osteogenesis. Chemotaxis of preosteoblasts toward future bone formation sites observed in the early stage of intramembranous osteogenesis is a critical cellular process for normal bone formation. However, molecular biological mechanisms of the chemotaxis of preosteoblasts are not fully understood. We have recently clarified, for the first time, the critical role of the cellular communication network factor 2 (CCN2)/connective tissue growth factor (CTGF)-integrin α5-Ras axis for chemotaxis of preosteoblasts during new bone formation through intramembranous osteogenesis. In this chapter, we describe in detail the procedures of the in vivo and in vitro assays to investigate the chemotactic property of CCN2/CTGF and its underlying molecular biological mechanisms during intramembranous osteogenesis.
Collapse
Affiliation(s)
- Nobuo Takeshita
- Section of Orthodontics and Dentofacial Orthopedics, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Miyagi, Japan
| | - Teruko Takano-Yamamoto
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Miyagi, Japan.
- Department of Biomaterials and Bioengineering, Faculty of Dental Medicine, Hokkaido University, Sapporo, Hokkaido, Japan.
| |
Collapse
|
69
|
Kadoya Y, Futaki S, Shimono C, Kimura T, Sekiguchi K. Dynamics, structure and assembly of the basement membrane in developing salivary glands revealed by an exogenous EGFP-tagged nidogen probe. Microscopy (Oxf) 2022; 71:357-363. [PMID: 35950724 DOI: 10.1093/jmicro/dfac040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/25/2022] [Accepted: 08/10/2022] [Indexed: 12/13/2022] Open
Abstract
Most epithelial tissues rapidly become complex during embryonic development while being surrounded by the basement membrane (BM). Thus, the BM shape is thought to change dramatically as the epithelium grows, but the underlying mechanism is not yet clear. Nidogen-1 is ubiquitous in the BM and binds to various other BM components, including laminin and type IV collagen. To elucidate the behavior of the BM during epithelial morphogenesis, we attempted to live-label the developing BM with recombinant human nidogen-1 fused to an enhanced green fluorescent protein (hNid1-EGFP). Submandibular glands of mouse embryos were cultured in glass-bottomed dishes and incubated in media containing hNid1-EGFP. Subsequent confocal microscopy clearly visualized the BMs surrounding the epithelial end buds. On three-dimensional reconstruction from Z-series confocal sections, the epithelial BM was observed as a thin sheet that expanded continuously around the entire epithelial basal surface. Because the explants continued to grow well in the presence of hNid1-EGFP, time-lapse confocal microscopy was performed to follow the dynamics of the BM. We found that the epithelial BM is an adaptive structure that deforms in accordance with the rapid shape changes of the developing epithelium. Furthermore, hNid1-EGFP was found to be incorporated differently into the epithelial BM compared with that reported for fibronectin or type IV collagen, suggesting that individual BM components assemble in different ways to form the BM.
Collapse
Affiliation(s)
- Yuichi Kadoya
- Laboratory of Anatomical Science, Kitasato University School of Allied Health Sciences, 1-15-1, Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
- Regenerative Medicine and Cell Design Research Facility, Kitasato University School of Allied Health Sciences, 1-15-1, Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Sugiko Futaki
- Department of Anatomy, Faculty of Medicine, Osaka Medical and Pharmaceutical University, 2-7, Daigakumachi, Takatsuki, Osaka 569-8686, Japan
- Division of Extracelluar Matrix Biochemistry, Institute for Protein Research, Osaka University, 3-2, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Chisei Shimono
- Division of Extracelluar Matrix Biochemistry, Institute for Protein Research, Osaka University, 3-2, Yamadaoka, Suita, Osaka 565-0871, Japan
- Nippi Research Institute of Biomatrix, Nippi Inc., 520-11, Kuwabara, Toride, Ibaraki 302-0017, Japan
| | - Taketoshi Kimura
- Laboratory of Anatomical Science, Kitasato University School of Allied Health Sciences, 1-15-1, Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
- Regenerative Medicine and Cell Design Research Facility, Kitasato University School of Allied Health Sciences, 1-15-1, Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Kiyotoshi Sekiguchi
- Division of Extracelluar Matrix Biochemistry, Institute for Protein Research, Osaka University, 3-2, Yamadaoka, Suita, Osaka 565-0871, Japan
- Division of Matrixome Research and Application, Institute for Protein Research, Osaka University, 3-2, Yamadaoka, Suita, Osaka 565-0871, Osaka, Japan
| |
Collapse
|
70
|
Guo F, Huang Y, Fernando T, Shi Y. Altered Molecular Pathways and Biomarkers of Endometrial Receptivity in Infertile Women with Polycystic Ovary Syndrome. Reprod Sci 2022; 29:3335-3345. [PMID: 35006579 DOI: 10.1007/s43032-022-00845-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 12/31/2021] [Indexed: 12/14/2022]
Abstract
Anovulation is the most prominent cause of infertility in polycystic ovary syndrome (PCOS) patients. Although ovulation can be corrected pharmacologically, the number of pregnancies remains low. Even if excellent embryos are transferred by IVF, it does not change the high miscarriage rate of PCOS patients. These facts collectively indicate that there is a disorder of endometrial development and receptivity to the embryo in PCOS patients, including the decrease of receptive ability, inhibition of embryo adhesion, undersupply of energy, poor blood perfusion, and pro-inflammatory status in the endometrium. However, it has never received the same attention as ovulatory dysfunction. Here we list some alternations of endometrial receptivity in women with PCOS, discuss the underlying intricate mechanisms, and try to find out the possible therapeutic targets, which may bring new perspectives to those who are able to provide high-quality embryos.
Collapse
Affiliation(s)
- Fei Guo
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, China
| | - Yufan Huang
- Department of Pharmacy, Mindong Hospital, Fujian Medical University, Ningde, 355000, Fujian, China
| | - Taniya Fernando
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, China
| | - Yingli Shi
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, China.
| |
Collapse
|
71
|
Abstract
Renal fibrosis is a hallmark of end-stage chronic kidney disease. It is characterized by increased accumulation of extracellular matrix (ECM), which disrupts cellular organization and function within the kidney. Here, we review the bi-directional interactions between cells and the ECM that drive renal fibrosis. We will discuss the cells involved in renal fibrosis, changes that occur in the ECM, the interactions between renal cells and the surrounding fibrotic microenvironment, and signal transduction pathways that are misregulated as fibrosis proceeds. Understanding the underlying mechanisms of cell-ECM crosstalk will identify novel targets to better identify and treat renal fibrosis and associated renal disease.
Collapse
Affiliation(s)
- Kristin P. Kim
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Caitlin E. Williams
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Christopher A. Lemmon
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| |
Collapse
|
72
|
Chau TCY, Keyser MS, Da Silva JA, Morris EK, Yordanov TE, Duscyz KP, Paterson S, Yap AS, Hogan BM, Lagendijk AK. Dynamically regulated focal adhesions coordinate endothelial cell remodelling in developing vasculature. Development 2022; 149:285926. [PMID: 36314606 DOI: 10.1242/dev.200454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 10/20/2022] [Indexed: 12/13/2022]
Abstract
The assembly of a mature vascular network involves coordinated endothelial cell (EC) shape changes, including the process of EC elongation. How EC elongation is dynamically regulated in vivo is not fully understood. Here, we have generated a zebrafish mutant that is deficient for the integrin adaptor protein Talin 1 (Tln1). Using a new focal adhesion (FA) marker line expressing endothelial Vinculinb-eGFP, we demonstrate that EC FAs function dynamically and are lost in our tln1 mutants, allowing us to uncouple the primary roles of FAs in EC morphogenesis from the secondary effects that occur due to systemic vessel failure or loss of blood flow. Tln1 loss led to compromised F-actin rearrangements, perturbed EC elongation and disrupted cell-cell junction linearisation in vessel remodelling. Finally, chemical induction of actin polymerisation restored actin dynamics and EC elongation during vascular morphogenesis. Together, we identify that FAs are essential for EC elongation and junction linearisation in flow-pressured vessels and that they influence actin polymerisation in cellular morphogenesis. These observations can explain the severely compromised vessel beds and vascular leakage observed in mutant models that lack integrin signalling. This article has an associated 'The people behind the papers' interview.
Collapse
Affiliation(s)
- Tevin C Y Chau
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Mikaela S Keyser
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Jason A Da Silva
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Elysse K Morris
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Teodor E Yordanov
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Kinga P Duscyz
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Scott Paterson
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre and The PeterMac Callum Department of Oncology, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Alpha S Yap
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Benjamin M Hogan
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre and The PeterMac Callum Department of Oncology, The University of Melbourne, Melbourne, Victoria 3000, Australia.,Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Anne Karine Lagendijk
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia.,School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
73
|
Ha BH, Yigit S, Natarajan N, Morse EM, Calderwood DA, Boggon TJ. Molecular basis for integrin adhesion receptor binding to p21-activated kinase 4 (PAK4). Commun Biol 2022; 5:1257. [PMID: 36385162 PMCID: PMC9669019 DOI: 10.1038/s42003-022-04157-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 10/24/2022] [Indexed: 11/18/2022] Open
Abstract
Integrin adhesion receptors provide links between extracellular ligands and cytoplasmic signaling. Multiple kinases have been found to directly engage with integrin β tails, but the molecular basis for these interactions remain unknown. Here, we assess the interaction between the kinase domain of p21-activated kinase 4 (PAK4) and the cytoplasmic tail of integrin β5. We determine three crystal structures of PAK4-β5 integrin complexes and identify the PAK-binding site. This is a region in the membrane-proximal half of the β5 tail and confirmed by site-directed mutagenesis. The β5 tail engages the kinase substrate-binding groove and positions the non-phosphorylatable integrin residue Glu767 at the phosphoacceptor site. Consistent with this, integrin β5 is poorly phosphorylated by PAK4, and in keeping with its ability to occlude the substrate-binding site, weakly inhibits kinase activity. These findings demonstrate the molecular basis for β5 integrin-PAK4 interactions but suggest modifications in understanding the potential cellular role of this interaction.
Collapse
Affiliation(s)
- Byung Hak Ha
- The Department of Pharmacology, Yale University, 333 Cedar St., New Haven, CT, 06520, USA
| | - Sezin Yigit
- The Department of Pharmacology, Yale University, 333 Cedar St., New Haven, CT, 06520, USA
| | - Nalini Natarajan
- The Department of Pharmacology, Yale University, 333 Cedar St., New Haven, CT, 06520, USA
| | - Elizabeth M Morse
- The Department of Cell Biology, Yale University, 333 Cedar St., New Haven, CT, 06520, USA
| | - David A Calderwood
- The Department of Pharmacology, Yale University, 333 Cedar St., New Haven, CT, 06520, USA.
- The Department of Cell Biology, Yale University, 333 Cedar St., New Haven, CT, 06520, USA.
| | - Titus J Boggon
- The Department of Pharmacology, Yale University, 333 Cedar St., New Haven, CT, 06520, USA.
- The Department of Molecular Biophysics and Biochemistry, Yale University, 333 Cedar St., New Haven, CT, 06520, USA.
| |
Collapse
|
74
|
Sklenářová R, Akla N, Latorre MJ, Ulrichová J, Franková J. Collagen as a Biomaterial for Skin and Corneal Wound Healing. J Funct Biomater 2022; 13:jfb13040249. [PMID: 36412890 PMCID: PMC9680244 DOI: 10.3390/jfb13040249] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/11/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022] Open
Abstract
The cornea and the skin are two organs that form the outer barrier of the human body. When either is injured (e.g., from surgery, physical trauma, or chemical burns), wound healing is initiated to restore integrity. Many cells are activated during wound healing. In particular, fibroblasts that are stimulated often transition into repair fibroblasts or myofibroblasts that synthesize extracellular matrix (ECM) components into the wound area. Control of wound ECM deposition is critical, as a disorganized ECM can block restoration of function. One of the most abundant structural proteins in the mammalian ECM is collagen. Collagen type I is the main component in connective tissues. It can be readily obtained and purified, and short analogs have also been developed for tissue engineering applications, including modulating the wound healing response. This review discusses the effect of several current collagen implants on the stimulation of corneal and skin wound healing. These range from collagen sponges and hydrogels to films and membranes.
Collapse
Affiliation(s)
- Renáta Sklenářová
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacký University in Olomouc, 775 15 Olomouc, Czech Republic
- Maisonneuve-Rosemont Hospital Research Centre, Montréal, QC H1T 2M4, Canada
| | - Naoufal Akla
- Maisonneuve-Rosemont Hospital Research Centre, Montréal, QC H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | | | - Jitka Ulrichová
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacký University in Olomouc, 775 15 Olomouc, Czech Republic
| | - Jana Franková
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacký University in Olomouc, 775 15 Olomouc, Czech Republic
- Correspondence:
| |
Collapse
|
75
|
Li R, Frangogiannis NG. Integrins in cardiac fibrosis. J Mol Cell Cardiol 2022; 172:1-13. [PMID: 35872324 DOI: 10.1016/j.yjmcc.2022.07.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/10/2022] [Accepted: 07/18/2022] [Indexed: 12/14/2022]
Abstract
Cells sense mechanical stress and changes in their matrix environment through the integrins, a family of heterodimeric surface receptors that bind to extracellular matrix ligands and trigger cytoskeletal remodeling, while transducing a wide range of intracellular signals. Integrins have been extensively implicated in regulation of inflammation, repair and fibrosis in many different tissues. This review manuscript discusses the role of integrin-mediated cascades in myocardial fibrosis. In vitro studies have demonstrated that β1 and αv integrins play an important role in fibrogenic conversion of cardiac fibroblast, acting through direct stimulation of FAK/Src cascades, or via accentuation of growth factor signaling. Fibrogenic actions of αv integrins may be mediated, at least in part, through pericellular activation of latent TGF-β stores. In vivo evidence supporting the role of integrin heterodimers in fibrotic cardiac remodeling is limited to associative evidence, and to experiments using pharmacologic inhibitors, or global loss-of-function approaches. Studies documenting in vivo actions of integrins on fibroblasts using cell-specific strategies are lacking. Integrin effects on leukocytes may also contribute to the pathogenesis of fibrotic myocardial responses by mediating recruitment and activation of fibrogenic macrophages. The profile and role of integrins in cardiac fibrosis may be dependent on the underlying pathologic condition. Considering their cell surface localization and the availability of small molecule inhibitors, integrins may be attractive therapeutic targets for patients with heart failure associated with prominent fibrotic remodeling.
Collapse
Affiliation(s)
- Ruoshui Li
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, United States of America.
| |
Collapse
|
76
|
Nguyen TH, Dao HH, Duong CM, Nguyen XH, Hoang DH, Do XH, Truong TQ, Nguyen TD, Nguyen LT, Than UTT. Cytokine-primed umbilical cord mesenchymal stem cells enhanced therapeutic effects of extracellular vesicles on osteoarthritic chondrocytes. Front Immunol 2022; 13:1041592. [PMID: 36389838 PMCID: PMC9647019 DOI: 10.3389/fimmu.2022.1041592] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 10/13/2022] [Indexed: 11/29/2022] Open
Abstract
In recent years, extracellular vesicles (EVs) secreted by mesenchymal stem cells (MSCs) have emerged as a potential cell-free therapy against osteoarthritis (OA). Thus, we investigated the therapeutic effects of EVs released by cytokine-primed umbilical cord-derived MSCs (UCMSCs) on osteoarthritic chondrocyte physiology. Priming UCMSCs individually with transforming growth factor beta (TGFβ), interferon alpha (IFNα), or tumor necrosis factor alpha (TNFα) significantly reduced the sorting of miR-181b-3p but not miR-320a-3p; two negative regulators of chondrocyte regeneration, into EVs. However, the EV treatment did not show any significant effect on chondrocyte proliferation. Meanwhile, EVs from both non-priming and cytokine-primed UCMSCs induced migration at later time points of measurement. Moreover, TGFβ-primed UCMSCs secreted EVs that could upregulate the expression of chondrogenesis markers (COL2 and ACAN) and downregulate fibrotic markers (COL1 and RUNX2) in chondrocytes. Hence, priming UCMSCs with cytokines can deliver selective therapeutic effects of EV treatment in OA and chondrocyte-related disorders.
Collapse
Affiliation(s)
- Thu Huyen Nguyen
- Center of Applied Sciences, Regenerative Medicine and Advance Technologies, Vinmec Healthcare System, Hanoi, Vietnam
| | - Huy Hoang Dao
- Center of Applied Sciences, Regenerative Medicine and Advance Technologies, Vinmec Healthcare System, Hanoi, Vietnam
- Faculty of Biology, VNU University of Science, Vietnam National University, Hanoi, Vietnam
| | - Chau Minh Duong
- Center of Applied Sciences, Regenerative Medicine and Advance Technologies, Vinmec Healthcare System, Hanoi, Vietnam
- Department of Biology, Clark University, Worcester, MA, United States
| | - Xuan-Hung Nguyen
- Center of Applied Sciences, Regenerative Medicine and Advance Technologies, Vinmec Healthcare System, Hanoi, Vietnam
- College of Health Sciences, VinUniversity, Hanoi, Vietnam
| | - Diem Huong Hoang
- Center of Applied Sciences, Regenerative Medicine and Advance Technologies, Vinmec Healthcare System, Hanoi, Vietnam
| | - Xuan-Hai Do
- Department of Practical and Experimental Surgery, Vietnam Military Medical University, Hanoi, Vietnam
| | - Trung Quang Truong
- Hanoi Medical University, Hanoi Medical University Hospital, Hanoi, Vietnam
| | - Tu Dac Nguyen
- Center of Applied Sciences, Regenerative Medicine and Advance Technologies, Vinmec Healthcare System, Hanoi, Vietnam
| | - Liem Thanh Nguyen
- College of Health Sciences, VinUniversity, Hanoi, Vietnam
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Uyen Thi Trang Than
- Center of Applied Sciences, Regenerative Medicine and Advance Technologies, Vinmec Healthcare System, Hanoi, Vietnam
- *Correspondence: Uyen Thi Trang Than,
| |
Collapse
|
77
|
Role of Vitronectin and Its Receptors in Neuronal Function and Neurodegenerative Diseases. Int J Mol Sci 2022; 23:ijms232012387. [PMID: 36293243 PMCID: PMC9604229 DOI: 10.3390/ijms232012387] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022] Open
Abstract
Vitronectin (VTN), a multifunctional glycoprotein with various physiological functions, exists in plasma and the extracellular matrix. It is known to be involved in the cell attachment, spreading and migration through binding to the integrin receptor, mainly via the RGD sequence. VTN is also widely used in the maintenance and expansion of pluripotent stem cells, but its effects go beyond that. Recent evidence shows more functions of VTN in the nervous system as it participates in neural differentiation, neuronutrition and neurogenesis, as well as in regulating axon size, supporting and guiding neurite extension. Furthermore, VTN was proved to play a key role in protecting the brain as it can reduce the permeability of the blood-brain barrier by interacting with integrin receptors in vascular endothelial cells. Moreover, evidence suggests that VTN is associated with neurodegenerative diseases, such as Alzheimer's disease, but its function has not been fully understood. This review summarizes the functions of VTN and its receptors in neurons and describes the role of VTN in the blood-brain barrier and neurodegenerative diseases.
Collapse
|
78
|
Zhao X, Guo J, Jia X, Yang Y, Liu L, Nie W, Fang Z. Internalization of Leptospira interrogans via diverse endocytosis mechanisms in human macrophages and vascular endothelial cells. PLoS Negl Trop Dis 2022; 16:e0010778. [PMID: 36137081 PMCID: PMC9531806 DOI: 10.1371/journal.pntd.0010778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 10/04/2022] [Accepted: 08/31/2022] [Indexed: 11/18/2022] Open
Abstract
Leptospirosis, one of the leading global causes of morbidity and mortality, is an emerging public health problem, particularly in large urban centers of developing countries. Leptospirosis results from infection with an organism belonging to the Leptospira genus L. interrogans. The extensive invasive ability has previously been documented, however a mechanism that describes how the organism is internalized by human macrophages and transmigrates through human blood vessel remains poorly understood. In the present study, we utilized a human macrophage and vascular endothelial cell line to study the diverse invasive mechanisms by which L. interrogans infections occur. We found that THP-1 and HUVEC had a diverse expression of cell receptors and L. interrogans entered THP-1 and HUVEC by different pathways. In the macrophage model cell line, ITGB1/FAK-signaling mediated microfilament dependent endocytosis with lysosome fusion, whereas ITGB1/CAV-1/PI3K-signaling mediated microfilament dependent endocytosis and transcytosis without lysosome fusion in the endothelial cell model. Shedding of pathogenic leptospires from HUVEC displayed higher viability than those from THP-1. The monolayer of HUVEC maintained integrity during the infection, while 3D imaging showed that leptospires were transmigrated both intra- and intercellularly. These results indicate that endocytosis of leptospires in human macrophages and human vascular endothelial cells are quite different, macrophages are responsible for eliminating leptospires in the human body during the infection while vascular endothelial cells facilitate dissemination of leptospires from blood vessels into target organs where they cause injury. Leptospirosis is a zoonotic bacterial disease which causes 1.03 million cases and 58,900 deaths each year. Human infections occur when the primary reservoir hosts, such as rodents, contaminate food and water with leptospires. Unlike other bacterial pathogens, leptospires invade the human body through mucosal barriers and enter the bloodstream, which can result in septicemia. Left untreated, leptospirosis can spread into multiple organs and tissues such as lungs, liver and kidneys. Pathological features of this disease include high fever, myalgia, lymphadenectasis, hemorrhaging and jaundice. Human macrophages and vascular endothelial cells play important roles in eliminating and preventing the transmission of this pathogen. We speculated that leptospires could be neutralized in macrophages and subsequently transported by vascular endothelial cells throughout the human body. In this research, the diverse mechanisms of human macrophages and vascular endothelial cells infected by leptospires were explored. Our findings can be used to improve the treatment, prevention, and supervision regarding leptospire transmission and infection.
Collapse
Affiliation(s)
- Xin Zhao
- Institute of Health Quarantine, Chinese Academy of Inspection and Quarantine, Beijing, China
- * E-mail: (XZ); (ZF)
| | - Jun Guo
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Quality and Health of Tianjin, Tianjin University of Science & Technology, Tianjin, China
| | - Xiaoyuan Jia
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yaling Yang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Quality and Health of Tianjin, Tianjin University of Science & Technology, Tianjin, China
| | - Lijuan Liu
- Institute of Health Quarantine, Chinese Academy of Inspection and Quarantine, Beijing, China
| | - Weizhong Nie
- Department of health quarantine, Qinhuangdao Customs District, Qinhuangdao, China
| | - Zhiqiang Fang
- Institute of Health Quarantine, Chinese Academy of Inspection and Quarantine, Beijing, China
- * E-mail: (XZ); (ZF)
| |
Collapse
|
79
|
Bioimaging Nucleic-Acid Aptamers with Different Specificities in Human Glioblastoma Tissues Highlights Tumoral Heterogeneity. Pharmaceutics 2022; 14:pharmaceutics14101980. [PMID: 36297416 PMCID: PMC9609998 DOI: 10.3390/pharmaceutics14101980] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/07/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Nucleic-acid aptamers are of strong interest for diagnosis and therapy. Compared with antibodies, they are smaller, stable upon variations in temperature, easy to modify, and have higher tissue-penetration abilities. However, they have been little described as detection probes in histology studies of human tissue sections. In this study, we performed fluorescence imaging with two aptamers targeting cell-surface receptors EGFR and integrin α5β1, both involved in the aggressiveness of glioblastoma. The aptamers’ cell-binding specificities were confirmed using confocal imaging. The affinities of aptamers for glioblastoma cells expressing these receptors were in the 100–300 nM range. The two aptamers were then used to detect EGFR and integrin α5β1 in human glioblastoma tissues and compared with antibody labeling. Our aptafluorescence assays proved to be able to very easily reveal, in a one-step process, not only inter-tumoral glioblastoma heterogeneity (differences observed at the population level) but also intra-tumoral heterogeneity (differences among cells within individual tumors) when aptamers with different specificities were used simultaneously in multiplexing labeling experiments. The discussion also addresses the strengths and limitations of nucleic-acid aptamers for biomarker detection in histology.
Collapse
|
80
|
Abstract
Single-pass transmembrane receptors (SPTMRs) represent a diverse group of integral membrane proteins that are involved in many essential cellular processes, including signal transduction, cell adhesion, and transmembrane transport of materials. Dysregulation of the SPTMRs is linked with many human diseases. Despite extensive efforts in past decades, the mechanisms of action of the SPTMRs remain incompletely understood. One major hurdle is the lack of structures of the full-length SPTMRs in different functional states. Such structural information is difficult to obtain by traditional structural biology methods such as X-ray crystallography and nuclear magnetic resonance (NMR). The recent rapid development of single-particle cryo-electron microscopy (cryo-EM) has led to an exponential surge in the number of high-resolution structures of integral membrane proteins, including SPTMRs. Cryo-EM structures of SPTMRs solved in the past few years have tremendously improved our understanding of how SPTMRs function. In this review, we will highlight these progresses in the structural studies of SPTMRs by single-particle cryo-EM, analyze important structural details of each protein involved, and discuss their implications on the underlying mechanisms. Finally, we also briefly discuss remaining challenges and exciting opportunities in the field.
Collapse
Affiliation(s)
- Kai Cai
- Departments of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75231, USA
| | - Xuewu Zhang
- Departments of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75231, USA
- Departments of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75231, USA
- Corresponding Author: Xuewu Zhang, Department of pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Xiao-chen Bai
- Departments of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75231, USA
- Departments of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75231, USA
- Corresponding Author: Xiao-chen Bai, Department of Biophysics, UT Southwestern Medical Center, Dallas, TX 75390, USA;
| |
Collapse
|
81
|
Integrin β1/Cell Surface GRP78 Complex Regulates TGFβ1 and Its Profibrotic Effects in Response to High Glucose. Biomedicines 2022; 10:biomedicines10092247. [PMID: 36140347 PMCID: PMC9496450 DOI: 10.3390/biomedicines10092247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/07/2022] [Accepted: 09/07/2022] [Indexed: 11/17/2022] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of kidney failure worldwide. Characterized by overproduction and accumulation of extracellular matrix (ECM) proteins, glomerular sclerosis is its earliest manifestation. High glucose (HG) plays a central role by increasing matrix production by glomerular mesangial cells (MC). We previously showed that HG induces translocation of GRP78 from the endoplasmic reticulum to the cell surface (csGRP78), where it acts as a signaling molecule to promote intracellular profibrotic FAK/Akt activation. Here, we identify integrin β1 as a key transmembrane signaling partner for csGRP78. We show that it is required for csGRP78-regulated FAK/Akt activation in response to HG, as well as downstream production, secretion and activity of the well characterized profibrotic cytokine transforming growth factor β1 (TGFβ1). Intriguingly, integrin β1 also itself promotes csGRP78 translocation. Furthermore, integrin β1 effects on cytoskeletal organization are not required for its function in csGRP78 translocation and signaling. These data together support an important pathologic role for csGRP78/integrin β1 in mediating key profibrotic responses to HG in kidney cells. Inhibition of their interaction will be further evaluated as a therapeutic target to limit fibrosis progression in DKD.
Collapse
|
82
|
Zhang Z, Dalan R, Hu Z, Wang JW, Chew NW, Poh KK, Tan RS, Soong TW, Dai Y, Ye L, Chen X. Reactive Oxygen Species Scavenging Nanomedicine for the Treatment of Ischemic Heart Disease. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2202169. [PMID: 35470476 DOI: 10.1002/adma.202202169] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/08/2022] [Indexed: 06/14/2023]
Abstract
Ischemic heart disease (IHD) is the leading cause of disability and mortality worldwide. Reactive oxygen species (ROS) have been shown to play key roles in the progression of diabetes, hypertension, and hypercholesterolemia, which are independent risk factors that lead to atherosclerosis and the development of IHD. Engineered biomaterial-based nanomedicines are under extensive investigation and exploration, serving as smart and multifunctional nanocarriers for synergistic therapeutic effect. Capitalizing on cell/molecule-targeting drug delivery, nanomedicines present enhanced specificity and safety with favorable pharmacokinetics and pharmacodynamics. Herein, the roles of ROS in both IHD and its risk factors are discussed, highlighting cardiovascular medications that have antioxidant properties, and summarizing the advantages, properties, and recent achievements of nanomedicines that have ROS scavenging capacity for the treatment of diabetes, hypertension, hypercholesterolemia, atherosclerosis, ischemia/reperfusion, and myocardial infarction. Finally, the current challenges of nanomedicines for ROS-scavenging treatment of IHD and possible future directions are discussed from a clinical perspective.
Collapse
Affiliation(s)
- Zhan Zhang
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, 999078, China
| | - Rinkoo Dalan
- Department of Endocrinology, Tan Tock Seng Hospital, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 408433, Singapore
| | - Zhenyu Hu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Jiong-Wei Wang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Department of Diagnostic Radiology and Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Nicholas Ws Chew
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, 119074, Singapore
| | - Kian-Keong Poh
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, 119074, Singapore
| | - Ru-San Tan
- Department of Cardiology, National Heart Centre Singapore, Singapore, 119609, Singapore
| | - Tuck Wah Soong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yunlu Dai
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macao, Taipa, Macau SAR, 999078, China
| | - Lei Ye
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology and Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Department of Chemical and Biomolecular Engineering and Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| |
Collapse
|
83
|
Lunasin as a Promising Plant-Derived Peptide for Cancer Therapy. Int J Mol Sci 2022; 23:ijms23179548. [PMID: 36076946 PMCID: PMC9455814 DOI: 10.3390/ijms23179548] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022] Open
Abstract
Cancer has become one of the main public health problems worldwide, demanding the development of new therapeutic agents that can help reduce mortality. Lunasin is a soybean peptide that has emerged as an attractive option because its preventive and therapeutic actions against cancer. In this review, we evaluated available research on lunasin’s structure and mechanism of action, which should be useful for the development of lunasin-based therapeutic products. We described data on its primary, secondary, tertiary, and possible quaternary structure, susceptibility to post-translational modifications, and structural stability. These characteristics are important for understanding drug activity and characterizing lunasin products. We also provided an overview of research on lunasin pharmacokinetics and safety. Studies examining lunasin’s mechanisms of action against cancer were reviewed, highlighting reported activities, and known molecular partners. Finally, we briefly discussed commercially available lunasin products and potential combination therapeutics.
Collapse
|
84
|
The IRE1α-XBP1s Arm of the Unfolded Protein Response Activates N-Glycosylation to Remodel the Subepithelial Basement Membrane in Paramyxovirus Infection. Int J Mol Sci 2022; 23:ijms23169000. [PMID: 36012265 PMCID: PMC9408905 DOI: 10.3390/ijms23169000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/29/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
Respiratory syncytial virus (RSV) causes severe lower respiratory tract infections (LRTI) associated with decreased pulmonary function, asthma, and allergy. Recently, we demonstrated that RSV induces the hexosamine biosynthetic pathway via the unfolded protein response (UPR), which is a pathway controlling protein glycosylation and secretion of the extracellular matrix (ECM). Because the presence of matrix metalloproteinases and matricellular growth factors (TGF) is associated with severe LRTI, we studied the effect of RSV on ECM remodeling and found that RSV enhances the deposition of fibronectin-rich ECM by small airway epithelial cells in a manner highly dependent on the inositol requiring kinase (IRE1α)–XBP1 arm of the UPR. To understand this effect comprehensively, we applied pharmacoproteomics to understand the effect of the UPR on N-glycosylation and ECM secretion in RSV infection. We observe that RSV induces N-glycosylation and the secretion of proteins related to ECM organization, secretion, or proteins integral to plasma membranes, such as integrins, laminins, collagens, and ECM-modifying enzymes, in an IRE1α–XBP1 dependent manner. Using a murine paramyxovirus model that activates the UPR in vivo, we validate the IRE1α–XBP1-dependent secretion of ECM to alveolar space. This study extends understanding of the IRE1α–XBP1 pathway in regulating N-glycosylation coupled to structural remodeling of the epithelial basement membrane in RSV infection.
Collapse
|
85
|
Nagasaki K, Gavrilova O, Hajishengallis G, Somerman MJ. Does the RGD region of certain proteins affect metabolic activity? FRONTIERS IN DENTAL MEDICINE 2022. [DOI: 10.3389/fdmed.2022.974862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
A better understanding of the role of mineralized tissues and their associated factors in governing whole-body metabolism should be of value toward informing clinical strategies to treat mineralized tissue and metabolic disorders, such as diabetes and obesity. This perspective provides evidence suggesting a role for the arginine-glycine-aspartic acid (RGD) region, a sequence identified in several proteins secreted by bone cells, as well as other cells, in modulating systemic metabolic activity. We focus on (a) two of the SIBLING (small integrin-binding ligand, N-linked glycoprotein) family genes/proteins, bone sialoprotein (BSP) and osteopontin (OPN), (b) insulin-like growth factor-binding protein-1 & 2 (IGFBP-1, IGFBP-2) and (c) developmental endothelial locus 1 (DEL1) and milk fat globule–EGF factor-8 (MFG-E8). In addition, for our readers to appreciate the mounting evidence that a multitude of bone secreted factors affect the activity of other tissues, we provide a brief overview of other proteins, to include fibroblast growth factor 23 (FGF23), phosphatase orphan 1 (PHOSPHO1), osteocalcin (OCN/BGLAP), tissue non-specific alkaline phosphatase (TNAP) and acidic serine aspartic-rich MEPE-associated motif (ASARM), along with known/suggested functions of these factors in influencing energy metabolism.
Collapse
|
86
|
Cell Adhesion Strength Indicates the Antithrombogenicity of Poly(2-Methoxyethyl Acrylate) (PMEA): Potential Candidate for Artificial Small-Diameter Blood Vessel. SURFACES 2022. [DOI: 10.3390/surfaces5030027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Poly (2-methoxyethyl acrylate) (PMEA) is a US FDA-approved biocompatible polymer, although there is insufficient work on human umbilical vein endothelial cells (HUVECs) and platelet interaction analysis on PMEA-analogous polymers. In this study, we extensively investigated HUVEC–polymer and platelet–polymer interaction behavior by measuring the adhesion strength using single-cell force spectroscopy. Furthermore, the hydration layer of the polymer interface was observed using frequency-modulation atomic force microscopy. We found that endothelial cells can attach and spread on the PMEA surface with strong adhesion strength compared to other analogous polymers. We found that the hydration layers on the PMEA-analogous polymers were closely related to their weak platelet adhesion behavior. Based on our results, it can be concluded that PMEA is a promising candidate for the construction of artificial small-diameter blood vessels owing to the presence of IW and a hydration layer on the interface.
Collapse
|
87
|
Bonin F, Chiche A, Tariq Z, Azorin P, Nola S, Lidereau R, Driouch K. Kindlin-1 drives early steps of breast cancer metastasis. CANCER COMMUNICATIONS (LONDON, ENGLAND) 2022; 42:1036-1040. [PMID: 35881673 PMCID: PMC9558683 DOI: 10.1002/cac2.12338] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 05/20/2022] [Accepted: 07/08/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Florian Bonin
- Genetics Department, Institut Curie, Paris, 75005, France
| | - Aurélie Chiche
- Cell Biology and Cancer Department/Unit 144, Institut Curie/Centre National de Recherche Scientifique, Paris, 75005, France.,Department of Developmental & Stem Cell Biology/ Unit 3738, Institut Pasteur/Centre National de Recherche Scientifique, Paris, 75015, France
| | - Zakia Tariq
- Genetics Department, Institut Curie, Paris, 75005, France
| | - Paula Azorin
- Genetics Department, Institut Curie, Paris, 75005, France
| | - Sébastien Nola
- Genetics Department, Institut Curie, Paris, 75005, France.,Membrane Traffic in Health And Brain Disease /Unit 1266, Institute of Psychiatry and Neuroscience of Paris/Institut National de la Santé et de la Recherche Médicale, Université de Paris, Paris, 75014, France
| | | | | |
Collapse
|
88
|
Mechanical Properties of the Extracellular Environment of Human Brain Cells Drive the Effectiveness of Drugs in Fighting Central Nervous System Cancers. Brain Sci 2022; 12:brainsci12070927. [PMID: 35884733 PMCID: PMC9313046 DOI: 10.3390/brainsci12070927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/11/2022] [Accepted: 07/13/2022] [Indexed: 12/04/2022] Open
Abstract
The evaluation of nanomechanical properties of tissues in health and disease is of increasing interest to scientists. It has been confirmed that these properties, determined in part by the composition of the extracellular matrix, significantly affect tissue physiology and the biological behavior of cells, mainly in terms of their adhesion, mobility, or ability to mutate. Importantly, pathophysiological changes that determine disease development within the tissue usually result in significant changes in tissue mechanics that might potentially affect the drug efficacy, which is important from the perspective of development of new therapeutics, since most of the currently used in vitro experimental models for drug testing do not account for these properties. Here, we provide a summary of the current understanding of how the mechanical properties of brain tissue change in pathological conditions, and how the activity of the therapeutic agents is linked to this mechanical state.
Collapse
|
89
|
da Silva-Oliveira RJ, Gomes INF, da Silva LS, Lengert AVH, Laus AC, Melendez ME, Munari CC, Cury FDP, Longato GB, Reis RM. Efficacy of Combined Use of Everolimus and Second-Generation Pan-EGRF Inhibitors in KRAS Mutant Non-Small Cell Lung Cancer Cell Lines. Int J Mol Sci 2022; 23:ijms23147774. [PMID: 35887120 PMCID: PMC9317664 DOI: 10.3390/ijms23147774] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/04/2022] [Accepted: 07/11/2022] [Indexed: 01/27/2023] Open
Abstract
Background: EGFR mutations are present in approximately 15−50% of non-small cell lung cancer (NSCLC), which are predictive of anti-EGFR therapies. At variance, NSCLC patients harboring KRAS mutations are resistant to those anti-EGFR approaches. Afatinib and allitinib are second-generation pan-EGFR drugs, yet no predictive biomarkers are known in the NSCLC context. In the present study, we evaluated the efficacy of pan-EGFR inhibitors in a panel of 15 lung cancer cell lines associated with the KRAS mutations phenotype. Methods: KRAS wild-type sensitive NCI-H292 cell line was further transfected with KRAS mutations (p.G12D and p.G12S). The pan-EGFR inhibitors’ activity and biologic effect of KRAS mutations were evaluated by cytotoxicity, MAPK phospho-protein array, colony formation, migration, invasion, and adhesion. In addition, in vivo chicken chorioallantoic membrane assay was performed in KRAS mutant cell lines. The gene expression profile was evaluated by NanoString. Lastly, everolimus and pan-EGFR combinations were performed to determine the combination index. Results: The GI50 score classified two cell lines treated with afatinib and seven treated with allitinib as high-sensitive phenotypes. All KRAS mutant cell lines demonstrated a resistant profile for both therapies (GI50 < 30%). The protein array of KRAS edited cells indicated a significant increase in AKT, CREB, HSP27, JNK, and, importantly, mTOR protein levels compared with KRAS wild-type cells. The colony formation, migration, invasion, adhesion, tumor perimeter, and mesenchymal phenotype were increased in the H292 KRAS mutated cells. Gene expression analysis showed 18 dysregulated genes associated with the focal adhesion-PI3K-Akt-mTOR-signaling correlated in KRAS mutant cell lines. Moreover, mTOR overexpression in KRAS mutant H292 cells was inhibited after everolimus exposure, and sensitivity to afatinib and allitinib was restored. Conclusions: Our results indicate that allitinib was more effective than afatinib in NSCLC cell lines. KRAS mutations increased aggressive behavior through upregulation of the focal adhesion-PI3K-Akt-mTOR-signaling in NSCLC cells. Significantly, everolimus restored sensibility and improved cytotoxicity of EGFR inhibitors in the KRAS mutant NSCLC cell lines.
Collapse
Affiliation(s)
- Renato José da Silva-Oliveira
- Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (L.S.d.S.); (A.v.H.L.); (A.C.L.); (M.E.M.); (C.C.M.); (F.d.P.C.); (G.B.L.)
- Correspondence: (R.J.d.S.-O.); (R.M.R.)
| | - Izabela Natalia Faria Gomes
- Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (L.S.d.S.); (A.v.H.L.); (A.C.L.); (M.E.M.); (C.C.M.); (F.d.P.C.); (G.B.L.)
| | - Luciane Sussuchi da Silva
- Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (L.S.d.S.); (A.v.H.L.); (A.C.L.); (M.E.M.); (C.C.M.); (F.d.P.C.); (G.B.L.)
| | - André van Helvoort Lengert
- Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (L.S.d.S.); (A.v.H.L.); (A.C.L.); (M.E.M.); (C.C.M.); (F.d.P.C.); (G.B.L.)
| | - Ana Carolina Laus
- Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (L.S.d.S.); (A.v.H.L.); (A.C.L.); (M.E.M.); (C.C.M.); (F.d.P.C.); (G.B.L.)
| | - Matias Eliseo Melendez
- Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (L.S.d.S.); (A.v.H.L.); (A.C.L.); (M.E.M.); (C.C.M.); (F.d.P.C.); (G.B.L.)
| | - Carla Carolina Munari
- Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (L.S.d.S.); (A.v.H.L.); (A.C.L.); (M.E.M.); (C.C.M.); (F.d.P.C.); (G.B.L.)
| | - Fernanda de Paula Cury
- Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (L.S.d.S.); (A.v.H.L.); (A.C.L.); (M.E.M.); (C.C.M.); (F.d.P.C.); (G.B.L.)
| | - Giovanna Barbarini Longato
- Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (L.S.d.S.); (A.v.H.L.); (A.C.L.); (M.E.M.); (C.C.M.); (F.d.P.C.); (G.B.L.)
| | - Rui Manuel Reis
- Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (L.S.d.S.); (A.v.H.L.); (A.C.L.); (M.E.M.); (C.C.M.); (F.d.P.C.); (G.B.L.)
- Life and Health Sciences Research Institute (ICVS) Medical School, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga, Portugal
- Correspondence: (R.J.d.S.-O.); (R.M.R.)
| |
Collapse
|
90
|
Solubilization and Purification of α 5β 1 Integrin from Rat Liver for Reconstitution into Nanodiscs. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2507:1-18. [PMID: 35773574 DOI: 10.1007/978-1-0716-2368-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Transmembrane proteins (or integral membrane proteins) are synthesized in the endoplasmic reticulum where most of them are core glycosylated prior to folding and in some cases assembly into quaternary structures. Correctly glycosylated, folded, and assembled transmembrane proteins are then shuttled to the Golgi apparatus for additional posttranslational modifications such as complex-type glycosylations, sulfation or proteolytic clipping. At the plasma membrane, the glycosylated extracellular domains are key to communicate with the cellular environment for a variety of functions, such as binding to the extracellular matrix for cell adhesion and migration, to neighboring cells for cell-cell interaction, or to extracellular components for nutrient uptake and cell signaling. Intracellular domains are essential to mediate signaling cascades, or to connect to cytosolic adaptors for internalization and intracellular compartmentalization. Despite its importance for the understanding of molecular mechanisms of transmembrane protein function, the determination of their structures has remained a challenging task. In recent years, their reconstitution in lipid nanodiscs in combination with high resolution cryo-electron microscopy has provided novel avenues to render this process more accessible. Here, we describe detailed protocols for the solubilization of heavily glycosylated α5β1 integrin from rat livers, its purification and reconstitution into nanodiscs. At the plasma membrane of many cells, including tumor metastases, this essential heterodimeric transmembrane protein mediates the communication between extracellular matrix and cytosolic cytoskeleton in processes of cell adhesion and migration. We expect that the protocols that are described here will provide new opportunities for the determination of the full structure of α5β1 integrin, as well as for the understanding of how interacting partners can regulate function and activity of this transmembrane protein.
Collapse
|
91
|
Brock J, Erhardt J, Eisler SA, Hörning M. Optimization of Mechanosensitive Cross-Talk between Matrix Stiffness and Protein Density: Independent Matrix Properties Regulate Spreading Dynamics of Myocytes. Cells 2022; 11:2122. [PMID: 35805206 PMCID: PMC9265304 DOI: 10.3390/cells11132122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/29/2022] [Accepted: 07/02/2022] [Indexed: 11/25/2022] Open
Abstract
Cells actively sense differences in topology, matrix elasticity and protein composition of the extracellular microenvironment and adapt their function and morphology. In this study, we focus on the cross-talk between matrix stiffness and protein coating density that regulates morphology and proliferation dynamics of single myocytes. For this, C2C12 myocytes were monitored on L-DOPA functionalized hydrogels of 22 different elasticity and fibronectin density compositions. Static images were recorded and statistically analyzed to determine morphological differences and to identify the optimized extracellular matrix (ECM). Using that information, selected ECMs were used to study the dynamics before and after cell proliferation by statistical comparison of distinct cell states. We observed a fibronectin-density-independent increase of the projected cell area until 12 kPa. Additionally, changes in fibronectin density led to an area that was optimum at about 2.6 μg/cm2, which was confirmed by independent F-actin analysis, revealing a maximum actin-filament-to-cell-area ratio of 7.5%. Proliferation evaluation showed an opposite correlation between cell spreading duration and speed to matrix elasticity and protein density, which did not affect cell-cycle duration. In summary, we identified an optimized ECM composition and found that independent matrix properties regulate distinct cell characteristics.
Collapse
Affiliation(s)
- Judith Brock
- Biobased Materials Laboratory, Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, 70569 Stuttgart, Germany; (J.B.); (J.E.)
| | - Julia Erhardt
- Biobased Materials Laboratory, Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, 70569 Stuttgart, Germany; (J.B.); (J.E.)
| | - Stephan A. Eisler
- Stuttgart Research Center Systems Biology (SRCSB), University of Stuttgart, 70569 Stuttgart, Germany;
| | - Marcel Hörning
- Biobased Materials Laboratory, Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, 70569 Stuttgart, Germany; (J.B.); (J.E.)
| |
Collapse
|
92
|
Gerassimov N, Crain C, Bilyeu C, Jacob A, Fan CM. Examining the lineage autonomous role of β3-integrin in muscle regeneration. FASEB J 2022; 36:e22385. [PMID: 35734962 PMCID: PMC9236161 DOI: 10.1096/fj.202200464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/19/2022] [Indexed: 11/23/2022]
Abstract
Skeletal muscles can regenerate over the lifetime from resident muscle stem cells (MuSCs). Interactions between MuSCs and extracellular matrix (ECM) proteins are essential for muscle regeneration. The best‐known receptors for ECM proteins are integrins, a family composed of twenty‐some heterodimeric combinations of an α‐ and a β‐subunit. β1‐integrin (encoded by Itgb1) is required for quiescence, proliferation, migration, and fusion of Pax7+ MuSCs in the mouse model. β3‐integrin (encoded by Itgb3) has been reported to be critical for the myogenic differentiation of C2C12 myoblasts, and Itgb3 germline mutant mice were shown to regenerate few if any myofibers after injury. To investigate the autonomous role of Itgb3 in the myogenic lineage in vivo, we conditionally inactivated a floxed Itgb3 allele (Itgb3F) by constitutive Pax7‐Cre and tamoxifen‐inducible Pax7‐CreERT2 drivers. Unexpectedly, we found no defects in muscle regeneration in both conditional knockout models. In vitro studies using Itgb3 mutant myoblasts or RNAi knockdown of Itgb3 in myoblasts also did not reveal a role for myogenic differentiation. As β1‐ and β3‐integrins share ECM ligands and downstream signaling effectors, we further examined Itgb3's role in a Itgb1 haploid background. Still, we found no evidence for an autonomous role of Itgb3 in muscle regeneration in vivo. Thus, while Itgb3 is critical for the differentiation of C2C12 cells, the regenerative defects reported for the Itgb3 germline mutant are not due to its role in the MuSC. We conclude that if β3‐integrin does have a role in Pax7+ MuSCs, it is compensated by β1‐ and/or another β‐integrin(s).
Collapse
Affiliation(s)
- Nathalie Gerassimov
- Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland, USA
| | - Colt Crain
- Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland, USA.,Department of Cell, Molecular and Developmental Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Colin Bilyeu
- Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland, USA
| | | | - Chen-Ming Fan
- Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland, USA.,Department of Cell, Molecular and Developmental Biology, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
93
|
Ruiz-Fernández AR, Campos L, Gutierrez-Maldonado SE, Núñez G, Villanelo F, Perez-Acle T. Nanosecond Pulsed Electric Field (nsPEF): Opening the Biotechnological Pandora’s Box. Int J Mol Sci 2022; 23:ijms23116158. [PMID: 35682837 PMCID: PMC9181413 DOI: 10.3390/ijms23116158] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/23/2022] [Accepted: 05/23/2022] [Indexed: 02/04/2023] Open
Abstract
Nanosecond Pulsed Electric Field (nsPEF) is an electrostimulation technique first developed in 1995; nsPEF requires the delivery of a series of pulses of high electric fields in the order of nanoseconds into biological tissues or cells. They primary effects in cells is the formation of membrane nanopores and the activation of ionic channels, leading to an incremental increase in cytoplasmic Ca2+ concentration, which triggers a signaling cascade producing a variety of effects: from apoptosis up to cell differentiation and proliferation. Further, nsPEF may affect organelles, making nsPEF a unique tool to manipulate and study cells. This technique is exploited in a broad spectrum of applications, such as: sterilization in the food industry, seed germination, anti-parasitic effects, wound healing, increased immune response, activation of neurons and myocites, cell proliferation, cellular phenotype manipulation, modulation of gene expression, and as a novel cancer treatment. This review thoroughly explores both nsPEF’s history and applications, with emphasis on the cellular effects from a biophysics perspective, highlighting the role of ionic channels as a mechanistic driver of the increase in cytoplasmic Ca2+ concentration.
Collapse
Affiliation(s)
- Alvaro R. Ruiz-Fernández
- Computational Biology Lab, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Santiago 7780272, Chile; (L.C.); (S.E.G.-M.); (G.N.); (F.V.)
- Facultad de Ingeniería y Tecnología, Universidad San Sebastian, Bellavista 7, Santiago 8420524, Chile
- Correspondence: (A.R.R.-F.); (T.P.-A.)
| | - Leonardo Campos
- Computational Biology Lab, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Santiago 7780272, Chile; (L.C.); (S.E.G.-M.); (G.N.); (F.V.)
- Facultad de Ingeniería y Tecnología, Universidad San Sebastian, Bellavista 7, Santiago 8420524, Chile
| | - Sebastian E. Gutierrez-Maldonado
- Computational Biology Lab, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Santiago 7780272, Chile; (L.C.); (S.E.G.-M.); (G.N.); (F.V.)
- Facultad de Ingeniería y Tecnología, Universidad San Sebastian, Bellavista 7, Santiago 8420524, Chile
| | - Gonzalo Núñez
- Computational Biology Lab, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Santiago 7780272, Chile; (L.C.); (S.E.G.-M.); (G.N.); (F.V.)
| | - Felipe Villanelo
- Computational Biology Lab, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Santiago 7780272, Chile; (L.C.); (S.E.G.-M.); (G.N.); (F.V.)
- Facultad de Ingeniería y Tecnología, Universidad San Sebastian, Bellavista 7, Santiago 8420524, Chile
| | - Tomas Perez-Acle
- Computational Biology Lab, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Santiago 7780272, Chile; (L.C.); (S.E.G.-M.); (G.N.); (F.V.)
- Facultad de Ingeniería y Tecnología, Universidad San Sebastian, Bellavista 7, Santiago 8420524, Chile
- Correspondence: (A.R.R.-F.); (T.P.-A.)
| |
Collapse
|
94
|
Murase H, El-Sheikh Ali H, Ruby RE, Scoggin KE, Ball BA. Transcriptomic analysis of the chorioallantois in equine premature placental separation. Equine Vet J 2022; 55:405-418. [PMID: 35622344 DOI: 10.1111/evj.13602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 05/12/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Equine premature placental separation (PPS) is poorly understood and represents an important risk factor for fetal/neonatal hypoxia. OBJECTIVES To examine transcriptomic changes in the chorioallantois (CA) from mares with clinical PPS compared to the CA from normal foaling mares. Differential gene expression was determined and gene ontology as well as molecular pathways related to PPS were characterised. STUDY DESIGN Retrospective case: control study. METHODS CA were collected from Thoroughbred mares with a clinical history of PPS (n=33) and from control Thoroughbred mares (n=4) with normal parturition for examination of transcriptional changes in the placenta associated with PPS. Transcriptomic changes in the villous CA near the cervical star were determined by Illumina® sequencing and subsequent bioinformatic analysis. PPS samples were divided by k-means clustering, and differentially expressed genes (DEGs) in each PPS cluster were identified by comparing to controls. Shared DEGs between PPS clusters were used for gene ontology analysis and pathway analysis. RESULTS A total of 1204 DEGs were identified between PPS and control. Gene ontology revealed extracellular matrix (ECM) and cell adhesion, and pathway analysis revealed fatty acid, p-53, hypoxia, and inflammation. Eleven key regulator genes of PPS including growth factors (IGF1, TGFB2, TGFB3), transcription factors (HIF1A, JUNB, SMAD3), and transmembrane receptors (FGFR1, TNFRSF1A, TYROBP) were also identified. MAIN LIMITATIONS The use of clinical history of PPS, in the absence of other criteria, may have led to misidentification of some cases as PPS. CONCLUSIONS Transcriptomic analysis indicated that changes in ECM and cell adhesion were important factors in equine PPS. Key predicted upstream events include genes associated with hypoxia, inflammation and growth factors related to the pathogenesis of equine PPS.
Collapse
Affiliation(s)
- Harutaka Murase
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, USA.,Equine Science Division, Hidaka Training and Research Center, Japan Racing Association, Urakawa, Hokkaido, Japan
| | - Hossam El-Sheikh Ali
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, USA.,Faculty of Veterinary Medicine, Mansoura University, Egypt
| | - Rebecca E Ruby
- Veterinary Diagnostic Laboratory, University of Kentucky, Lexington, KY, USA
| | - Kirsten E Scoggin
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| | - Barry A Ball
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
95
|
Qin L, He T, Yang D, Wang Y, Li Z, Yan Q, Zhang P, Chen Z, Lin S, Gao H, Yao Q, Xu Z, Tang B, Yi W, Xiao G. Osteocyte β1 integrin loss causes low bone mass and impairs bone mechanotransduction in mice. J Orthop Translat 2022; 34:60-72. [PMID: 35615639 PMCID: PMC9119859 DOI: 10.1016/j.jot.2022.03.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/19/2022] [Accepted: 03/21/2022] [Indexed: 11/09/2022] Open
Abstract
Background The key focal adhesion protein β1 integrin plays an essential role in early skeletal development. However, roles of β1 integrin expression in osteocytes during the regulation of bone homeostasis and mechanotransduction are incompletely understood. Materials and methods To study the in vivo function of osteocyte β1 integrin in bone, we utilized the 10-kb Dmp1 (Dentin matrix acidic phosphoprotein 1)-Cre to generate mice with β1 integrin deletion in this cell type. Micro-computerized tomography, bone histomorphometry and immunohistochemistry were performed to determine the effects of osteocyte β1 integrin loss on bone mass accrual and biomechanical properties. In vivo tibial loading model was applied to study the possible involvement of osteocyte β1 integrin in bone mechanotransduction. Results Loss of β1 integrin expression in osteocytes resulted in a severe low bone mass and impaired biomechanical properties in load-bearing long bones and spines, but not in non-weight-bearing calvariae, in mice. The loss of β1 integrin led to enlarged size of lacunar-canalicular system, abnormal cell morphology, and disorientated nuclei in osteocytes. Furthermore, β1 integrin loss caused shortening and disorientated collagen I fibers in long bones. Osteocyte β1 integrin loss did not impact the osteoclast activities, but significantly reduced the osteoblast bone formation rate and, in the meantime, enhanced the adipogenic differentiation of the bone marrow stromal cells in the bone microenvironment. In addition, tibial loading failed to accelerate the anabolic bone formation and improve collagen I fiber integrity in mutant mice. Conclusions Our studies demonstrate an essential role of osteocyte β1 integrin in regulating bone homeostasis and mechanotransduction. The transnational potential of this article : This study reveals the regulatory roles of osteocyte β1 integrin in vivo for the maintenance of bone mass accrual, biomechanical properties, extracellular matrix integrity as well as bone mechanobiology, which defines β1 integrin a potential therapeutic target for skeletal diseases, such as osteoporosis.
Collapse
Affiliation(s)
- Lei Qin
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518000, China
| | - Tailin He
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
| | - Dazhi Yang
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518000, China
| | - Yishu Wang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
| | - Zhenjian Li
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Qinnan Yan
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
| | - Peijun Zhang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
| | - Zecai Chen
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518000, China
| | - Sixiong Lin
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
- Department of Spine Surgery, Orthopedic Research Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, 510080, China
| | - Huanqing Gao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
| | - Qing Yao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
| | - Zhen Xu
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518000, China
| | - Bin Tang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Weihong Yi
- Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518000, China
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
| |
Collapse
|
96
|
Guenther C. β2-Integrins - Regulatory and Executive Bridges in the Signaling Network Controlling Leukocyte Trafficking and Migration. Front Immunol 2022; 13:809590. [PMID: 35529883 PMCID: PMC9072638 DOI: 10.3389/fimmu.2022.809590] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 03/11/2022] [Indexed: 12/12/2022] Open
Abstract
Leukocyte trafficking is an essential process of immunity, occurring as leukocytes travel within the bloodstream and as leukocyte migration within tissues. While it is now established that leukocytes can utilize the mesenchymal migration mode or amoeboid migration mode, differences in the migratory behavior of leukocyte subclasses and how these are realized on a molecular level in each subclass is not fully understood. To outline these differences, first migration modes and their dependence on parameters of the extracellular environments will be explained, as well as the intracellular molecular machinery that powers migration in general. Extracellular parameters are detected by adhesion receptors such as integrins. β2-integrins are surface receptors exclusively expressed on leukocytes and are essential for leukocytes exiting the bloodstream, as well as in mesenchymal migration modes, however, integrins are dispensable for the amoeboid migration mode. Additionally, the balance of different RhoGTPases - which are downstream of surface receptor signaling, including integrins - mediate formation of membrane structures as well as actin dynamics. Individual leukocyte subpopulations have been shown to express distinct RhoGTPase profiles along with their differences in migration behavior, which will be outlined. Emerging aspects of leukocyte migration include signal transduction from integrins via actin to the nucleus that regulates DNA status, gene expression profiles and ultimately leukocyte migratory phenotypes, as well as altered leukocyte migration in tumors, which will be touched upon.
Collapse
Affiliation(s)
- Carla Guenther
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| |
Collapse
|
97
|
Sasaki H, Sasaki N. Tensin 2-deficient nephropathy - mechanosensitive nephropathy, genetic susceptibility. Exp Anim 2022; 71:252-263. [PMID: 35444113 PMCID: PMC9388341 DOI: 10.1538/expanim.22-0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Tensin 2 (TNS2), a focal adhesion protein, is considered to anchor focal adhesion proteins to β integrin as an integrin adaptor protein and/or serve as a scaffold to facilitate the
interactions of these proteins. In the kidney, TNS2 localizes to the basolateral surface of glomerular epithelial cells, i.e., podocytes. Loss of TNS2 leads to the development of glomerular
basement membrane lesions and abnormal accumulation of extracellular matrix in maturing glomeruli during the early postnatal stages. It subsequently results in podocyte foot process
effacement, eventually leading to glomerulosclerosis. Histopathological features of the affected glomeruli in the middle stage of the disease include expansion of the mesangial matrix
without mesangial cell proliferation. In this review, we provide an overview of TNS2-deficient nephropathy and discuss the potential mechanism underlying this mechanosensitive nephropathy,
which may be applicable to other glomerulonephropathies, such as CD151-deficient nephropathy and Alport syndrome. The onset of TNS2-deficient nephropathy strictly depends on the genetic
background, indicating the presence of critical modifier genes. A better understanding of molecular mechanisms of mechanosensitive nephropathy may open new avenues for the management of
patients with glomerulonephropathies.
Collapse
Affiliation(s)
- Hayato Sasaki
- Laboratory of Laboratory Animal Science and Medicine, School of Veterinary Medicine, Kitasato University
| | - Nobuya Sasaki
- Laboratory of Laboratory Animal Science and Medicine, School of Veterinary Medicine, Kitasato University
| |
Collapse
|
98
|
Chiang HY, Chu PH, Chen SC, Lee TH. MFG-E8 promotes osteogenic transdifferentiation of smooth muscle cells and vascular calcification by regulating TGF-β1 signaling. Commun Biol 2022; 5:364. [PMID: 35440618 PMCID: PMC9018696 DOI: 10.1038/s42003-022-03313-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 03/24/2022] [Indexed: 11/23/2022] Open
Abstract
Vascular calcification occurs in arterial aging, atherosclerosis, diabetes mellitus, and chronic kidney disease. Transforming growth factor-β1 (TGF-β1) is a key modulator driving the osteogenic transdifferentiation of vascular smooth muscle cells (VSMCs), leading to vascular calcification. We hypothesize that milk fat globule–epidermal growth factor 8 (MFG-E8), a glycoprotein expressed in VSMCs, promotes the osteogenic transdifferentiation of VSMCs through the activation of TGF-β1-mediated signaling. We observe that the genetic deletion of MFG-E8 prevents calcium chloride-induced vascular calcification in common carotid arteries (CCAs). The exogenous application of MFG-E8 to aged CCAs promotes arterial wall calcification. MFG-E8-deficient cultured VSMCs exhibit decreased biomineralization and phenotypic transformation to osteoblast-like cells in response to osteogenic medium. MFG-E8 promotes β1 integrin–dependent MMP2 expression, causing TGF-β1 activation and subsequent VSMC osteogenic transdifferentiation and biomineralization. Thus, the established molecular link between MFG-E8 and vascular calcification suggests that MFG-E8 can be therapeutically targeted to mitigate vascular calcification. A molecular link between the milk fat globule–epidermal growth factor 8 (MFG-E8), activation of vascular calcification driver TGF-β1 and osteogenic differentiation of vascular smooth muscle cells suggests that MFG-E8 could be a therapeutic target for vascular calcification.
Collapse
Affiliation(s)
- Hou-Yu Chiang
- Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Pao-Hsien Chu
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Shao-Chi Chen
- Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ting-Hein Lee
- Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan. .,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan. .,Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan.
| |
Collapse
|
99
|
Norris EG, Pan XS, Hocking DC. Receptor binding domain of SARS-CoV-2 is a functional αv-integrin agonist. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.04.11.487882. [PMID: 35441172 PMCID: PMC9016641 DOI: 10.1101/2022.04.11.487882] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Among the novel mutations distinguishing SARS-CoV-2 from similar respiratory coronaviruses is a K403R substitution in the receptor-binding domain (RBD) of the viral spike (S) protein within its S1 region. This amino acid substitution occurs near the angiotensin-converting enzyme 2 (ACE2)-binding interface and gives rise to a canonical RGD adhesion motif that is often found in native extracellular matrix proteins, including fibronectin. In the present study, the ability of recombinant S1-RBD to bind to cell surface integrins and trigger downstream signaling pathways was assessed and compared to RGD-containing, integrin-binding fragments of fibronectin. S1-RBD supported adhesion of both fibronectin-null mouse embryonic fibroblasts as well as primary human small airway epithelial cells. Cell adhesion to S1-RBD was cation- and RGD-dependent, and was inhibited by blocking antibodies against α v and β 3 , but not α 5 or β 1 , integrins. Similarly, direct binding of S1-RBD to recombinant human α v β 3 and α v β 6 integrins, but not α 5 β 1 integrins, was observed by surface plasmon resonance. Adhesion to S1-RBD initiated cell spreading, focal adhesion formation, and actin stress fiber organization to a similar extent as fibronectin. Moreover, S1-RBD stimulated tyrosine phosphorylation of the adhesion mediators FAK, Src, and paxillin, Akt activation, and supported cell proliferation. Together, these data demonstrate that the RGD sequence within S1-RBD can function as an α v -selective integrin agonist. This study provides evidence that cell surface α v -containing integrins can respond functionally to spike protein and raise the possibility that S1-mediated dysregulation of ECM dynamics may contribute to the pathogenesis and/or post-acute sequelae of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Emma G. Norris
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | - Xuan Sabrina Pan
- Department of Biomedical Engineering University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | - Denise C. Hocking
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
- Department of Biomedical Engineering University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| |
Collapse
|
100
|
Rosmus DD, Lange C, Ludwig F, Ajami B, Wieghofer P. The Role of Osteopontin in Microglia Biology: Current Concepts and Future Perspectives. Biomedicines 2022; 10:biomedicines10040840. [PMID: 35453590 PMCID: PMC9027630 DOI: 10.3390/biomedicines10040840] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/26/2022] [Accepted: 03/27/2022] [Indexed: 12/14/2022] Open
Abstract
The innate immune landscape of the central nervous system (CNS), including the brain and the retina, consists of different myeloid cell populations with distinct tasks to fulfill. Whereas the CNS borders harbor extraparenchymal CNS-associated macrophages whose main duty is to build up a defense against invading pathogens and other damaging factors from the periphery, the resident immune cells of the CNS parenchyma and the retina, microglia, are highly dynamic cells with a plethora of functions during homeostasis and disease. Therefore, microglia are constantly sensing their environment and closely interacting with surrounding cells, which is in part mediated by soluble factors. One of these factors is Osteopontin (OPN), a multifunctional protein that is produced by different cell types in the CNS, including microglia, and is upregulated in neurodegenerative and neuroinflammatory conditions. In this review, we discuss the current literature about the interaction between microglia and OPN in homeostasis and several disease entities, including multiple sclerosis (MS), Alzheimer’s and cerebrovascular diseases (AD, CVD), amyotrophic lateral sclerosis (ALS), age-related macular degeneration (AMD) and diabetic retinopathy (DR), in the context of the molecular pathways involved in OPN signaling shaping the function of microglia. As nearly all CNS diseases are characterized by pathological alterations in microglial cells, accompanied by the disturbance of the homeostatic microglia phenotype, the emergence of disease-associated microglia (DAM) states and their interplay with factors shaping the DAM-signature, such as OPN, is of great interest for therapeutical interventions in the future.
Collapse
Affiliation(s)
| | - Clemens Lange
- Eye Center, Freiburg Medical Center, University of Freiburg, 79106 Freiburg, Germany; (C.L.); (F.L.)
- Ophtha-Lab, Department of Ophthalmology, St. Franziskus Hospital, 48145 Muenster, Germany
| | - Franziska Ludwig
- Eye Center, Freiburg Medical Center, University of Freiburg, 79106 Freiburg, Germany; (C.L.); (F.L.)
| | - Bahareh Ajami
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, OR 97239, USA;
| | - Peter Wieghofer
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany;
- Cellular Neuroanatomy, Institute of Theoretical Medicine, Medical Faculty, Augsburg University, 86159 Augsburg, Germany
- Correspondence:
| |
Collapse
|